1
|
Ligorio C, Martinez-Espuga M, Laurenza D, Hartley A, Rodgers CB, Kotowska AM, Scurr DJ, Dalby MJ, Ordóñez-Morán P, Mata A. Disassembly of self-assembling peptide hydrogels as a versatile method for cell extraction and manipulation. J Mater Chem B 2024. [PMID: 39449374 PMCID: PMC11502993 DOI: 10.1039/d4tb01575d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Accepted: 10/15/2024] [Indexed: 10/26/2024]
Abstract
Self-assembling peptide hydrogels (SAPHs) are increasingly being used as two-dimensional (2D) cell culture substrates and three-dimensional (3D) matrices due to their tunable properties and biomimicry of native tissues. Despite these advantages, SAPHs often represent an end-point in cell culture, as isolating cells from them leads to low yields and disruption of cells, limiting their use and post-culture analyses. Here, we report on a protocol designed to easily and effectively disassemble peptide amphiphile (PA) SAPHs to retrieve 3D encapsulated cells with high viability and minimal disruption. Due to the pivotal role played by salt ions in SAPH gelation, tetrasodium ethylenediaminetetraacetic acid (Na4EDTA) was used as metal chelator to sequester ions participating in PA self-assembly and induce a rapid, efficient, clean, and gentle gel-to-sol transition. We characterise PA disassembly from the nano- to the macro-scale, provide mechanistic and practical insights into the PA disassembly mechanism, and assess the potential use of the process. As proof-of-concept, we isolated different cell types from cell-laden PA hydrogels and demonstrated the possibility to perform downstream biological analyses including cell re-plating, gene analysis, and flow cytometry with high reproducibility and no material interference. Our work offers new opportunities for the use of SAPHs in cell culture and the potential use of cells cultured on SAPHs, in applications such as cell expansion, analysis of in vitro models, cell therapies, and regenerative medicine.
Collapse
Affiliation(s)
- Cosimo Ligorio
- Biodiscovery Institute, University of Nottingham, Nottingham, UK.
- School of Pharmacy, University of Nottingham, Nottingham, UK
- Department of Chemical and Environmental Engineering, University of Nottingham, Nottingham, UK
| | - Magda Martinez-Espuga
- Biodiscovery Institute, University of Nottingham, Nottingham, UK.
- School of Pharmacy, University of Nottingham, Nottingham, UK
| | - Domenico Laurenza
- Biodiscovery Institute, University of Nottingham, Nottingham, UK.
- School of Pharmacy, University of Nottingham, Nottingham, UK
| | - Alex Hartley
- Biodiscovery Institute, University of Nottingham, Nottingham, UK.
- School of Pharmacy, University of Nottingham, Nottingham, UK
| | - Chloe B Rodgers
- Centre for the Cellular Microenvironment, School of Molecular Biosciences, College of Medical, Veterinary and Life Sciences, Mazumdar-Shaw Advanced Research Centre, University of Glasgow, Glasgow G11 6EW, UK
| | - Anna M Kotowska
- School of Pharmacy, University of Nottingham, Nottingham, UK
| | - David J Scurr
- School of Pharmacy, University of Nottingham, Nottingham, UK
| | - Matthew J Dalby
- Centre for the Cellular Microenvironment, School of Molecular Biosciences, College of Medical, Veterinary and Life Sciences, Mazumdar-Shaw Advanced Research Centre, University of Glasgow, Glasgow G11 6EW, UK
| | - Paloma Ordóñez-Morán
- Translational Medical Sciences Unit, School of Medicine, Centre for Cancer Sciences, Biodiscovery Institute, University of Nottingham, Nottingham, UK
| | - Alvaro Mata
- Biodiscovery Institute, University of Nottingham, Nottingham, UK.
- School of Pharmacy, University of Nottingham, Nottingham, UK
- Department of Chemical and Environmental Engineering, University of Nottingham, Nottingham, UK
| |
Collapse
|
2
|
Demiray EB, Sezgin Arslan T, Derkus B, Arslan YE. A Facile Strategy for Preparing Flexible and Porous Hydrogel-Based Scaffolds from Silk Sericin/Wool Keratin by In Situ Bubble-Forming for Muscle Tissue Engineering Applications. Macromol Biosci 2024:e2400362. [PMID: 39427341 DOI: 10.1002/mabi.202400362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 09/15/2024] [Indexed: 10/22/2024]
Abstract
In the present study, it is aimed to fabricate a novel silk sericin (SS)/wool keratin (WK) hydrogel-based scaffolds using an in situ bubble-forming strategy containing an N-(3-dimethylaminopropyl)-N'-ethylcarbodiimide hydrochloride (EDC) and N-hydroxysuccinimide (NHS) coupling reaction. During the rapid gelation process, CO2 bubbles are released by activating the carboxyl groups in sericin with EDC and NHS, entrapped within the gel, creating a porous cross-linked structure. With this approach, five different hydrogels (S2K1, S4K2, S2K4, S6K3, and S3K6) are constructed to investigate the impact of varying sericin and keratin ratios. Analyses reveal that more sericin in the proteinaceous mixture reinforced the hydrogel network. Additionally, the hydrogels' pore size distribution, swelling ratio, wettability, and in vitro biodegradation rate, which are crucial for the applications of biomaterials, are evaluated. Moreover, biocompatibility and proangiogenic properties are analyzed using an in-ovo chorioallantoic membrane assay. The findings suggest that the S4K2 hydrogel exhibited the most promising characteristics, featuring an adequately flexible and highly porous structure. The results obtained by in vitro assessments demonstrate the potential of S4K2 hydrogel in muscle tissue engineering. However, further work is necessary to improve hydrogels with an aligned structure to meet the features that can fully replace muscle tissue for volumetric muscle loss regeneration.
Collapse
Affiliation(s)
- Elif Beyza Demiray
- Regenerative Biomaterials Laboratory, Department of Bioengineering, Faculty of Engineering, Çanakkale Onsekiz Mart University, Çanakkale, 17100, Turkey
| | - Tugba Sezgin Arslan
- Regenerative Biomaterials Laboratory, Department of Bioengineering, Faculty of Engineering, Çanakkale Onsekiz Mart University, Çanakkale, 17100, Turkey
| | - Burak Derkus
- Stem Cell Research Lab, Department of Chemistry, Faculty of Science, Ankara University, Ankara, 06100, Turkey
| | - Yavuz Emre Arslan
- Regenerative Biomaterials Laboratory, Department of Bioengineering, Faculty of Engineering, Çanakkale Onsekiz Mart University, Çanakkale, 17100, Turkey
| |
Collapse
|
3
|
Carbajo-Gordillo AI, Benito E, Galbis E, Grosso R, Iglesias N, Valencia C, Lucas R, García-Martín MG, de-Paz MV. Simultaneous Formation of Polyhydroxyurethanes and Multicomponent Semi-IPN Hydrogels. Polymers (Basel) 2024; 16:880. [PMID: 38611138 PMCID: PMC11013152 DOI: 10.3390/polym16070880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 03/16/2024] [Accepted: 03/19/2024] [Indexed: 04/14/2024] Open
Abstract
This study introduces an efficient strategy for synthesizing polyhydroxyurethane-based multicomponent hydrogels with enhanced rheological properties. In a single-step process, 3D materials composed of Polymer 1 (PHU) and Polymer 2 (PVA or gelatin) were produced. Polymer 1, a crosslinked polyhydroxyurethane (PHU), grew within a colloidal solution of Polymer 2, forming an interconnected network. The synthesis of Polymer 1 utilized a Non-Isocyanate Polyurethane (NIPU) methodology based on the aminolysis of bis(cyclic carbonate) (bisCC) monomers derived from 1-thioglycerol and 1,2-dithioglycerol (monomers A and E, respectively). This method, applied for the first time in Semi-Interpenetrating Network (SIPN) formation, demonstrated exceptional orthogonality since the functional groups in Polymer 2 do not interfere with Polymer 1 formation. Optimizing PHU formation involved a 20-trial methodology, identifying influential variables such as polymer concentration, temperature, solvent (an aprotic and a protic solvent), and the organo-catalyst used [a thiourea derivative (TU) and 1,8-diazabicyclo [5.4.0]undec-7-ene (DBU)]. The highest molecular weights were achieved under near-bulk polymerization conditions using TU-protic and DBU-aprotic as catalyst-solvent combinations. Monomer E-based PHU exhibited higher Mw¯ than monomer A-based PHU (34.1 kDa and 16.4 kDa, respectively). Applying the enhanced methodology to prepare 10 multicomponent hydrogels using PVA or gelatin as the polymer scaffold revealed superior rheological properties in PVA-based hydrogels, exhibiting solid-like gel behavior. Incorporating monomer E enhanced mechanical properties and elasticity (with loss tangent values of 0.09 and 0.14). SEM images unveiled distinct microstructures, including a sponge-like pattern in certain PVA-based hydrogels when monomer A was chosen, indicating the formation of highly superporous interpenetrated materials. In summary, this innovative approach presents a versatile methodology for obtaining advanced hydrogel-based systems with potential applications in various biomedical fields.
Collapse
Affiliation(s)
- Ana I. Carbajo-Gordillo
- Dpto. Química Orgánica y Farmacéutica, Facultad de Farmacia, Universidad de Sevilla, 41012 Sevilla, Spain (R.L.); (M.-G.G.-M.)
| | - Elena Benito
- Dpto. Química Orgánica y Farmacéutica, Facultad de Farmacia, Universidad de Sevilla, 41012 Sevilla, Spain (R.L.); (M.-G.G.-M.)
| | - Elsa Galbis
- Dpto. Química Orgánica y Farmacéutica, Facultad de Farmacia, Universidad de Sevilla, 41012 Sevilla, Spain (R.L.); (M.-G.G.-M.)
| | - Roberto Grosso
- Dpto. Química Orgánica y Farmacéutica, Facultad de Farmacia, Universidad de Sevilla, 41012 Sevilla, Spain (R.L.); (M.-G.G.-M.)
| | - Nieves Iglesias
- Dpto. Química Orgánica y Farmacéutica, Facultad de Farmacia, Universidad de Sevilla, 41012 Sevilla, Spain (R.L.); (M.-G.G.-M.)
| | - Concepción Valencia
- Dpto. Ingeniería Química, Facultad de Ciencias Experimentales, Campus El Carmen, Universidad de Huelva, 21071 Huelva, Spain
- Pro2TecS—Chemical Process and Product Technology Research Center, Universidad de Huelva, 21071 Huelva, Spain
| | - Ricardo Lucas
- Dpto. Química Orgánica y Farmacéutica, Facultad de Farmacia, Universidad de Sevilla, 41012 Sevilla, Spain (R.L.); (M.-G.G.-M.)
| | - M.-Gracia García-Martín
- Dpto. Química Orgánica y Farmacéutica, Facultad de Farmacia, Universidad de Sevilla, 41012 Sevilla, Spain (R.L.); (M.-G.G.-M.)
| | - M.-Violante de-Paz
- Dpto. Química Orgánica y Farmacéutica, Facultad de Farmacia, Universidad de Sevilla, 41012 Sevilla, Spain (R.L.); (M.-G.G.-M.)
| |
Collapse
|
4
|
Chen H, Xue H, Zeng H, Dai M, Tang C, Liu L. 3D printed scaffolds based on hyaluronic acid bioinks for tissue engineering: a review. Biomater Res 2023; 27:137. [PMID: 38142273 DOI: 10.1186/s40824-023-00460-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Accepted: 11/07/2023] [Indexed: 12/25/2023] Open
Abstract
Hyaluronic acid (HA) is widely distributed in human connective tissue, and its unique biological and physicochemical properties and ability to facilitate biological structure repair make it a promising candidate for three-dimensional (3D) bioprinting in the field of tissue regeneration and biomedical engineering. Moreover, HA is an ideal raw material for bioinks in tissue engineering because of its histocompatibility, non-immunogenicity, biodegradability, anti-inflammatory properties, anti-angiogenic properties, and modifiability. Tissue engineering is a multidisciplinary field focusing on in vitro reconstructions of mammalian tissues, such as cartilage tissue engineering, neural tissue engineering, skin tissue engineering, and other areas that require further clinical applications. In this review, we first describe the modification methods, cross-linking methods, and bioprinting strategies for HA and its derivatives as bioinks and then critically discuss the strengths, shortcomings, and feasibility of each method. Subsequently, we reviewed the practical clinical applications and outcomes of HA bioink in 3D bioprinting. Finally, we describe the challenges and opportunities in the development of HA bioink to provide further research references and insights.
Collapse
Affiliation(s)
- Han Chen
- The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325200, China
- Ningxia Medical University, Ningxia, 750004, China
- Xijing Hospital of Air Force Military Medical University, Xi'an, 710032, China
| | - Huaqian Xue
- The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325200, China
- Ningxia Medical University, Ningxia, 750004, China
| | - Huanxuan Zeng
- The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325200, China
| | - Minghai Dai
- The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325200, China
| | - Chengxuan Tang
- The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325200, China.
| | - Liangle Liu
- The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325200, China.
| |
Collapse
|
5
|
Isik M, Okesola BO, Eylem CC, Kocak E, Nemutlu E, D'Este M, Mata A, Derkus B. Bioactive and chemically defined hydrogels with tunable stiffness guide cerebral organoid formation and modulate multi-omics plasticity in cerebral organoids. Acta Biomater 2023; 171:223-238. [PMID: 37793600 DOI: 10.1016/j.actbio.2023.09.040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 09/20/2023] [Accepted: 09/25/2023] [Indexed: 10/06/2023]
Abstract
Organoids are an emerging technology with great potential in human disease modelling, drug development, diagnosis, tissue engineering, and regenerative medicine. Organoids as 3D-tissue culture systems have gained special attention in the past decades due to their ability to faithfully recapitulate the complexity of organ-specific tissues. Despite considerable successes in culturing physiologically relevant organoids, their real-life applications are currently limited by challenges such as scarcity of an appropriate biomimetic matrix. Peptide amphiphiles (PAs) due to their well-defined chemistry, tunable bioactivity, and extracellular matrix (ECM)-like nanofibrous architecture represent an attractive material scaffold for organoids development. Using cerebral organoids (COs) as exemplar, we demonstrate the possibility to create bio-instructive hydrogels with tunable stiffness ranging from 0.69 kPa to 2.24 kPa to culture and induce COs growth. We used orthogonal chemistry involving oxidative coupling and supramolecular interactions to create two-component hydrogels integrating the bio-instructive activity and ECM-like nanofibrous architecture of a laminin-mimetic PAs (IKVAV-PA) and tunable crosslinking density of hyaluronic acid functionalized with tyramine (HA-Try). Multi-omics technology including transcriptomics, proteomics, and metabolomics reveals the induction and growth of COs in soft HA-Tyr hydrogels containing PA-IKVAV such that the COs display morphology and biomolecular signatures similar to those grown in Matrigel scaffolds. Our materials hold great promise as a safe synthetic ECM for COs induction and growth. Our approach represents a well-defined alternative to animal-derived matrices for the culture of COs and might expand the applicability of organoids in basic and clinical research. STATEMENT OF SIGNIFICANCE: Synthetic bio-instructive materials which display tissue-specific functionality and nanoscale architecture of the native extracellular matrix are attractive matrices for organoids development. These synthetic matrices are chemically defined and animal-free compared to current gold standard matrices such as Matrigel. Here, we developed hydrogel matrices with tunable stiffness, which incorporate laminin-mimetic peptide amphiphiles to grow and expand cerebral organoids. Using multi-omics tools, the present study provides exciting data on the effects of neuro-inductive cues on the biomolecular profiles of brain organoids.
Collapse
Affiliation(s)
- Melis Isik
- Stem Cell Research Lab, Department of Chemistry, Faculty of Science, Ankara University, Ankara 06560, Turkey
| | - Babatunde O Okesola
- School of Life Sciences, Faculty of Medicine and Health Sciences, University of Nottingham, Nottingham NG7 2UH, UK
| | - Cemil Can Eylem
- Analytical Chemistry Division, Faculty of Pharmacy, Hacettepe University, Ankara 06230, Turkey
| | - Engin Kocak
- Division of Analytical Chemistry, Faculty of Gulhane Pharmacy, Health Science University, Ankara 06018, Turkey
| | - Emirhan Nemutlu
- Analytical Chemistry Division, Faculty of Pharmacy, Hacettepe University, Ankara 06230, Turkey; Bioanalytic and Omics Laboratory, Faculty of Pharmacy, Hacettepe University, Ankara, Turkey
| | - Matteo D'Este
- AO Research Institute Davos, Clavadelerstrasse 8, Davos Platz 7270, Switzerland
| | - Alvaro Mata
- School of Pharmacy University of Nottingham, University Park, Nottingham NG7 2RD, UK; Department of Chemical and Environmental Engineering, University of Nottingham, University Park, Nottingham NG7 2RD, UK
| | - Burak Derkus
- Stem Cell Research Lab, Department of Chemistry, Faculty of Science, Ankara University, Ankara 06560, Turkey.
| |
Collapse
|
6
|
Isik M, Karakaya E, Arslan TS, Atila D, Erdogan YK, Arslan YE, Eskizengin H, Eylem CC, Nemutlu E, Ercan B, D'Este M, Okesola BO, Derkus B. 3D Printing of Extracellular Matrix-Based Multicomponent, All-Natural, Highly Elastic, and Functional Materials toward Vascular Tissue Engineering. Adv Healthc Mater 2023; 12:e2203044. [PMID: 37014809 PMCID: PMC11468991 DOI: 10.1002/adhm.202203044] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 03/12/2023] [Indexed: 04/05/2023]
Abstract
3D printing offers an exciting opportunity to fabricate biological constructs with specific geometries, clinically relevant sizes, and functions for biomedical applications. However, successful application of 3D printing is limited by the narrow range of printable and bio-instructive materials. Multicomponent hydrogel bioinks present unique opportunities to create bio-instructive materials able to display high structural fidelity and fulfill the mechanical and functional requirements for in situ tissue engineering. Herein, 3D printable and perfusable multicomponent hydrogel constructs with high elasticity, self-recovery properties, excellent hydrodynamic performance, and improved bioactivity are reported. The materials' design strategy integrates fast gelation kinetics of sodium alginate (Alg), in situ crosslinking of tyramine-modified hyaluronic acid (HAT), and temperature-dependent self-assembly and biological functions of decellularized aorta (dAECM). Using extrusion-based printing approach, the capability to print the multicomponent hydrogel bioinks with high precision into a well-defined vascular constructs able to withstand flow and repetitive cyclic compressive loading, is demonstrated. Both in vitro and pre-clinical models are used to show the pro-angiogenic and anti-inflammatory properties of the multicomponent vascular constructs. This study presents a strategy to create new bioink whose functional properties are greater than the sum of their components and with potential applications in vascular tissue engineering and regenerative medicine.
Collapse
Affiliation(s)
- Melis Isik
- Stem Cell Research LabDepartment of ChemistryFaculty of ScienceAnkara UniversityAnkara06560Turkey
| | - Ece Karakaya
- Stem Cell Research LabDepartment of ChemistryFaculty of ScienceAnkara UniversityAnkara06560Turkey
| | - Tugba Sezgin Arslan
- Stem Cell Research LabDepartment of ChemistryFaculty of ScienceAnkara UniversityAnkara06560Turkey
| | - Deniz Atila
- Department of Engineering SciencesMiddle East Technical UniversityAnkara06800Turkey
- International Centre for Research on Innovative Bio‐based Materials (ICRI‐BioM)Lodz University of TechnologyLodz90924Poland
| | - Yasar Kemal Erdogan
- Biomedical Engineering ProgramMiddle East Technical UniversityAnkara06800Turkey
- Department of Biomedical EngineeringIsparta University of Applied ScienceIsparta32260Turkey
| | - Yavuz Emre Arslan
- Regenerative Biomaterials Laboratory, Department of BioengineeringFaculty of EngineeringCanakkale Onsekiz Mart UniversityCanakkale17100Turkey
| | - Hakan Eskizengin
- Department of BiologyFaculty of ScienceAnkara UniversityAnkara06560Turkey
| | - Cemil Can Eylem
- Analytical Chemistry DivisionFaculty of PharmacyHacettepe UniversityAnkara06230Turkey
| | - Emirhan Nemutlu
- Analytical Chemistry DivisionFaculty of PharmacyHacettepe UniversityAnkara06230Turkey
- Bioanalytic and Omics LaboratoryFaculty of PharmacyHacettepe UniversityAnkara06100Turkey
| | - Batur Ercan
- Biomedical Engineering ProgramMiddle East Technical UniversityAnkara06800Turkey
- Department of Metallurgical and Materials EngineeringMiddle East Technical UniversityAnkara06800Turkey
| | - Matteo D'Este
- AO Research Institute DavosClavadelerstrasse 8Davos Platz7270Switzerland
| | - Babatunde O. Okesola
- School of Life Sciences, Faculty of Medicine and Health SciencesUniversity of NottinghamUniversity ParkNottinghamNG7 2UHUK
| | - Burak Derkus
- Stem Cell Research LabDepartment of ChemistryFaculty of ScienceAnkara UniversityAnkara06560Turkey
| |
Collapse
|
7
|
Ligorio C, Mata A. Synthetic extracellular matrices with function-encoding peptides. NATURE REVIEWS BIOENGINEERING 2023; 1:1-19. [PMID: 37359773 PMCID: PMC10127181 DOI: 10.1038/s44222-023-00055-3] [Citation(s) in RCA: 25] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Accepted: 03/16/2023] [Indexed: 06/28/2023]
Abstract
The communication of cells with their surroundings is mostly encoded in the epitopes of structural and signalling proteins present in the extracellular matrix (ECM). These peptide epitopes can be incorporated in biomaterials to serve as function-encoding molecules to modulate cell-cell and cell-ECM interactions. In this Review, we discuss natural and synthetic peptide epitopes as molecular tools to bioengineer bioactive hydrogel materials. We present a library of functional peptide sequences that selectively communicate with cells and the ECM to coordinate biological processes, including epitopes that directly signal to cells, that bind ECM components that subsequently signal to cells, and that regulate ECM turnover. We highlight how these epitopes can be incorporated in different biomaterials as individual or multiple signals, working synergistically or additively. This molecular toolbox can be applied in the design of biomaterials aimed at regulating or controlling cellular and tissue function, repair and regeneration.
Collapse
Affiliation(s)
- Cosimo Ligorio
- Biodiscovery Institute, University of Nottingham, Nottingham, UK
- Department of Chemical and Environmental Engineering, University of Nottingham, Nottingham, UK
| | - Alvaro Mata
- Biodiscovery Institute, University of Nottingham, Nottingham, UK
- Department of Chemical and Environmental Engineering, University of Nottingham, Nottingham, UK
- School of Pharmacy, University of Nottingham, Nottingham, UK
| |
Collapse
|
8
|
Halperin-Sternfeld M, Pokhojaev A, Ghosh M, Rachmiel D, Kannan R, Grinberg I, Asher M, Aviv M, Ma PX, Binderman I, Sarig R, Adler-Abramovich L. Immunomodulatory fibrous hyaluronic acid-Fmoc-diphenylalanine-based hydrogel induces bone regeneration. J Clin Periodontol 2023; 50:200-219. [PMID: 36110056 PMCID: PMC10086858 DOI: 10.1111/jcpe.13725] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 08/30/2022] [Accepted: 09/09/2022] [Indexed: 01/18/2023]
Abstract
AIM To investigate the potential of an ultrashort aromatic peptide hydrogelator integrated with hyaluronic acid (HA) to serve as a scaffold for bone regeneration. MATERIALS AND METHODS Fluorenylmethyloxycarbonyl-diphenylalanine (FmocFF)/HA hydrogel was prepared and characterized using microscopy and rheology. Osteogenic differentiation of MC3T3-E1 preosteoblasts was investigated using Alizarin red, alkaline phosphatase and calcium deposition assays. In vivo, 5-mm-diameter calvarial critical-sized defects were prepared in 20 Sprague-Dawley rats and filled with either FmocFF/HA hydrogel, deproteinized bovine bone mineral, FmocFF/Alginate hydrogel or left unfilled. Eight weeks after implantation, histology and micro-computed tomography analyses were performed. Immunohistochemistry was performed in six rats to assess the hydrogel's immunomodulatory effect. RESULTS A nanofibrous FmocFF/HA hydrogel with a high storage modulus of 46 KPa was prepared. It supported osteogenic differentiation of MC3T3-E1 preosteoblasts and facilitated calcium deposition. In vivo, the hydrogel implantation resulted in approximately 93% bone restoration. It induced bone deposition not only around the margins, but also generated bony islets along the defect. Elongated M2 macrophages lining at the periosteum-hydrogel interface were observed 1 week after implantation. After 3 weeks, these macrophages were dispersed through the regenerating tissue surrounding the newly formed bone. CONCLUSIONS FmocFF/HA hydrogel can serve as a cell-free, biomimetic, immunomodulatory scaffold for bone regeneration.
Collapse
Affiliation(s)
- Michal Halperin-Sternfeld
- Department of Oral Biology, The Maurice and Gabriela Goldschleger School of Dental Medicine, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,The Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv, Israel.,The Center for the Physics and Chemistry of Living Systems, Tel Aviv University, Tel Aviv, Israel
| | - Ariel Pokhojaev
- Department of Oral Biology, The Maurice and Gabriela Goldschleger School of Dental Medicine, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Moumita Ghosh
- Department of Oral Biology, The Maurice and Gabriela Goldschleger School of Dental Medicine, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,The Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv, Israel.,The Center for the Physics and Chemistry of Living Systems, Tel Aviv University, Tel Aviv, Israel.,Department of Chemistry, Techno India University, Kolkata, West Bengal, India
| | - Dana Rachmiel
- Department of Oral Biology, The Maurice and Gabriela Goldschleger School of Dental Medicine, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,The Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv, Israel.,The Center for the Physics and Chemistry of Living Systems, Tel Aviv University, Tel Aviv, Israel
| | - Raha Kannan
- Department of Biologic and Materials Sciences, University of Michigan, Ann Arbor, Michigan, USA
| | - Itzhak Grinberg
- Department of Oral Biology, The Maurice and Gabriela Goldschleger School of Dental Medicine, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,The Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv, Israel.,The Center for the Physics and Chemistry of Living Systems, Tel Aviv University, Tel Aviv, Israel
| | - Moshe Asher
- Department of Oral Biology, The Maurice and Gabriela Goldschleger School of Dental Medicine, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Moran Aviv
- Department of Oral Biology, The Maurice and Gabriela Goldschleger School of Dental Medicine, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,The Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv, Israel.,The Center for the Physics and Chemistry of Living Systems, Tel Aviv University, Tel Aviv, Israel.,School of Mechanical Engineering, Afeka Tel Aviv Academic College of Engineering, Tel Aviv, Israel
| | - Peter X Ma
- Department of Biologic and Materials Sciences, University of Michigan, Ann Arbor, Michigan, USA
| | - Itzhak Binderman
- Department of Oral Biology, The Maurice and Gabriela Goldschleger School of Dental Medicine, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Rachel Sarig
- Department of Oral Biology, The Maurice and Gabriela Goldschleger School of Dental Medicine, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,The Dan David Center for Human Evolution and Biohistory Research, Tel Aviv University, Tel Aviv, Israel
| | - Lihi Adler-Abramovich
- Department of Oral Biology, The Maurice and Gabriela Goldschleger School of Dental Medicine, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,The Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv, Israel.,The Center for the Physics and Chemistry of Living Systems, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
9
|
Randle RI, Ginesi RE, Matsarskaia O, Schweins R, Draper ER. Process Dependent Complexity in Multicomponent Gels. Macromol Rapid Commun 2023; 44:e2200709. [PMID: 36177680 PMCID: PMC11475255 DOI: 10.1002/marc.202200709] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 09/20/2022] [Indexed: 11/06/2022]
Abstract
Mixing low molecular weight gelators (LMWGs) can be used to combine favorable properties of the individual components within a multifunctional gel. Such multicomponent systems are complex enough in themselves but the method of combining components is not commonly considered something to influence self-assembly. Herein, two multicomponent systems comprising of a naphthalene-based dipeptide hydrogelator and one of two modified naphthalene diimides (NDIs), one of which forms gels, and the other does not, are investigated. These systems are probed, examining the structures formed and their gel properties (when preparing a solution from either a mixed powder of both components or by mixing pre-formed solutions of each component) using rheology, small angle neutron scattering (SANS), and absorbance spectroscopy. It is found that by altering the method of preparation, it is can either induce self-sorting or co-assembly within the fibers formed that underpin the gel network.
Collapse
Affiliation(s)
- Rebecca I. Randle
- School of Chemistry, Joseph Black BuildingUniversity of GlasgowGlasgowG12 8QQUK
| | - Rebecca E. Ginesi
- School of Chemistry, Joseph Black BuildingUniversity of GlasgowGlasgowG12 8QQUK
| | - Olga Matsarskaia
- Institut Laue‐LangevinLarge Scale Structures Group71 Avenue des Martyrs, CS 20156Grenoble CEDEX 9F‐38042France
| | - Ralf Schweins
- Institut Laue‐LangevinLarge Scale Structures Group71 Avenue des Martyrs, CS 20156Grenoble CEDEX 9F‐38042France
| | - Emily R. Draper
- School of Chemistry, Joseph Black BuildingUniversity of GlasgowGlasgowG12 8QQUK
| |
Collapse
|
10
|
Application of Hydrogels as Three-Dimensional Bioprinting Ink for Tissue Engineering. Gels 2023; 9:gels9020088. [PMID: 36826258 PMCID: PMC9956898 DOI: 10.3390/gels9020088] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 01/15/2023] [Accepted: 01/18/2023] [Indexed: 01/22/2023] Open
Abstract
The use of three-dimensional bioprinting technology combined with the principle of tissue engineering is important for the construction of tissue or organ regeneration microenvironments. As a three-dimensional bioprinting ink, hydrogels need to be highly printable and provide a stiff and cell-friendly microenvironment. At present, hydrogels are used as bioprinting inks in tissue engineering. However, there is still a lack of summary of the latest 3D printing technology and the properties of hydrogel materials. In this paper, the materials commonly used as hydrogel bioinks; the advanced technologies including inkjet bioprinting, extrusion bioprinting, laser-assisted bioprinting, stereolithography bioprinting, suspension bioprinting, and digital 3D bioprinting technologies; printing characterization including printability and fidelity; biological properties, and the application fields of bioprinting hydrogels in bone tissue engineering, skin tissue engineering, cardiovascular tissue engineering are reviewed, and the current problems and future directions are prospected.
Collapse
|
11
|
Karakaya E, Erdogan YK, Arslan TS, Arslan YE, Odabas S, Ercan B, Emregul E, Derkus B. Decellularized Bone Extracellular Matrix-Coated Electrospun PBAT Microfibrous Membranes with Cell Instructive Ability and Improved Bone Tissue Forming Capacity. Macromol Biosci 2022; 22:e2200303. [PMID: 36129099 DOI: 10.1002/mabi.202200303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 08/23/2022] [Indexed: 01/15/2023]
Abstract
Current approaches to develop bone tissue engineering scaffolds have some limitations and shortcomings. They mainly suffer from combining mechanical stability and bioactivity on the same platform. Synthetic polymers are able to produce mechanically stable sturctures with fibrous morphology when they are electrospun, however, they cannot exhibit bioactivity, which is crucial for tissue engineering and regenerative medicine. One current strategy to bring bioactivity in synthetic materials is to combine extracellular matrix (ECM)-sourced materials with biologically inert synthetic materials. ECM-sourced materials without any modifications are mechanically unstable; therefore, reinforcing them with mechanically stable platforms is indispensable. In order to overcome this bifacial problem, we have demonstrated that poly(butylene adipate-co-terephthalate) (PBAT) electrospun microfibrous membranes can be successfully modified with decellularized bone ECM to endow fibers with bioactive hydrogel and mimic natural micro-features of the native bone tissue. The developed structures have been shown to support osteogenesis, confirmed by histochemical staining and gene expression studies. Furthermore, ECM-coated PBAT fibers, when they were aligned, supplied an improved level of osteogenesis. The strategy demonstrated can be adapted to any other tissues, and the emerging microfibrous, mechanically stable, and bioactive materials can find implications in the specific fields of tissue engineering and regenerative medicine.
Collapse
Affiliation(s)
- Ece Karakaya
- Personalized Medicine and Biosensing Research (PMBR) Laboratory, Department of Chemistry, Faculty of Science, Ankara University, Ankara, 06560, Turkey
| | - Yasar Kemal Erdogan
- Biomedical Engineering Program, Middle East Technical University, Ankara, 06800, Turkey.,Department of Biomedical Engineering, Isparta University of Applied Science, Isparta, 32260, Turkey
| | - Tugba Sezgin Arslan
- Personalized Medicine and Biosensing Research (PMBR) Laboratory, Department of Chemistry, Faculty of Science, Ankara University, Ankara, 06560, Turkey
| | - Yavuz Emre Arslan
- Regenerative Biomaterials Laboratory, Department of Bioengineering, Faculty of Engineering, Canakkale Onsekiz Mart University, Canakkale, 17100, Turkey
| | - Sedat Odabas
- Biomaterials and Tissue Engineering Laboratory (BteLAB), Department of Chemistry, Faculty of Science, Ankara University, Besevler, Ankara, 06560, Turkey
| | - Batur Ercan
- Biomedical Engineering Program, Middle East Technical University, Ankara, 06800, Turkey.,Department of Metallurgical and Materials Engineering, Middle East Technical University, Ankara, 06800, Turkey
| | - Emel Emregul
- Personalized Medicine and Biosensing Research (PMBR) Laboratory, Department of Chemistry, Faculty of Science, Ankara University, Ankara, 06560, Turkey
| | - Burak Derkus
- Stem Cell Research Lab, Department of Chemistry, Faculty of Science, Ankara University, Besevler, Ankara, 06560, Turkey
| |
Collapse
|
12
|
Isik M, Okesola BO, Eylem CC, Kocak E, Nemutlu E, Emregul E, D'Este M, Derkus B. Tuning the Cell-Adhesive Properties of Two-Component Hybrid Hydrogels to Modulate Cancer Cell Behavior, Metastasis, and Death Pathways. Biomacromolecules 2022; 23:4254-4267. [PMID: 36136959 PMCID: PMC9554906 DOI: 10.1021/acs.biomac.2c00733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Abstract
![]()
This work presents a polysaccharide
and protein-based
two-component
hybrid hydrogel integrating the cell-adhesive gelatin-tyramine (G-Tyr)
and nonadhesive hyaluronic acid-tyramine (HA-Tyr) through enzyme-mediated
oxidative coupling reaction. The resulting HA-Tyr/G-Tyr hydrogel reflects
the precise chemical and mechanical features of the cancer extracellular
matrix and is able to tune cancer cell adhesion upon switching the
component ratio. The cells form quasi-spheroids on HA-Tyr rich hydrogels,
while they tend to form an invasive monolayer culture on G-Tyr rich
hydrogels. The metastatic genotype of colorectal adenocarcinoma cells
(HT-29) increases on G-Tyr rich hydrogels which is driven by the material’s
adhesive property, and additionally confirmed by the suppressed gene
expressions of apoptosis and autophagy. On the other hand, HA-Tyr
rich hydrogels lead the cells to necrotic death via oxidative stress
in quasi-spheroids. This work demonstrates the ideality of HA-Tyr/G-Tyr
to modulate cancer cell adhesion, which also has potential in preventing
primary metastasis after onco-surgery, biomaterials-based cancer research,
and drug testing.
Collapse
Affiliation(s)
- Melis Isik
- Interdisciplinary Research Unit for Advanced Materials (INTRAM), Department of Chemistry, Faculty of Science, Ankara University, Ankara 06560, Turkey
| | - Babatunde O Okesola
- Department of Eye and Vision Science, Institute of Life Course and Medical Sciences, Faculty of Medicine, University of Liverpool, Liverpool L7 8TX, U.K.,School of Life Science, Faculty of Medicine and Health Sciences, University of Nottingham, Nottingham NG7 2RD, U.K
| | - Cemil Can Eylem
- Analytical Chemistry Division, Faculty of Pharmacy, Hacettepe University, Ankara 06230, Turkey
| | - Engin Kocak
- Division of Analytical Chemistry, Faculty of Gulhane Pharmacy, Health Science University, Ankara 06018, Turkey
| | - Emirhan Nemutlu
- Analytical Chemistry Division, Faculty of Pharmacy, Hacettepe University, Ankara 06230, Turkey.,Bioanalytic and Omics Laboratory, Faculty of Pharmacy, Hacettepe University, Ankara 06230, Turkey
| | - Emel Emregul
- Interdisciplinary Research Unit for Advanced Materials (INTRAM), Department of Chemistry, Faculty of Science, Ankara University, Ankara 06560, Turkey
| | - Matteo D'Este
- AO Research Institute Davos, Clavadelerstrasse 8, Davos Platz 7270, Switzerland
| | - Burak Derkus
- Interdisciplinary Research Unit for Advanced Materials (INTRAM), Department of Chemistry, Faculty of Science, Ankara University, Ankara 06560, Turkey.,Stem Cell Research Lab, Department of Chemistry, Faculty of Science, Ankara University, Ankara 06560, Turkey
| |
Collapse
|
13
|
Lou J, Mooney DJ. Chemical strategies to engineer hydrogels for cell culture. Nat Rev Chem 2022; 6:726-744. [PMID: 37117490 DOI: 10.1038/s41570-022-00420-7] [Citation(s) in RCA: 100] [Impact Index Per Article: 50.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/27/2022] [Indexed: 12/12/2022]
Abstract
Two-dimensional and three-dimensional cell culture systems are widely used for biological studies, and are the basis of the organoid, tissue engineering and organ-on-chip research fields in applications such as disease modelling and drug screening. The natural extracellular matrix of tissues, a complex scaffold with varying chemical and mechanical properties, has a critical role in regulating important cellular functions such as spreading, migration, proliferation and differentiation, as well as tissue morphogenesis. Hydrogels are biomaterials that are used in cell culture systems to imitate critical features of a natural extracellular matrix. Chemical strategies to synthesize and tailor the properties of these hydrogels in a controlled manner, and manipulate their biological functions in situ, have been developed. In this Review, we provide the rational design criteria for predictably engineering hydrogels to mimic the properties of the natural extracellular matrix. We highlight the advances in using biocompatible strategies to engineer hydrogels for cell culture along with recent developments to dynamically control the cellular environment by exploiting stimuli-responsive chemistries. Finally, future opportunities to engineer hydrogels are discussed, in which the development of novel chemical methods will probably have an important role.
Collapse
|
14
|
Kumar A, Sood A, Singhmar R, Mishra YK, Thakur VK, Han SS. Manufacturing functional hydrogels for inducing angiogenic-osteogenic coupled progressions in hard tissue repairs: prospects and challenges. Biomater Sci 2022; 10:5472-5497. [PMID: 35994005 DOI: 10.1039/d2bm00894g] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
In large bone defects, inadequate vascularization within the engineered constructs has been a major challenge in developing clinically impactful products. It is fairly determined that bone tissues and blood vessels are established concurrently throughout tissue repairs after an injury. Thus, the coupling of angiogenesis-osteogenesis is an essential course of action in bone tissue restoration. The manufacture of biomaterial-based scaffolds plays a decisive role in stimulating angiogenic and osteogenic progressions (instruction of neovascularization and bone mineralization). Bone hydrogels with optimal conditions are more efficient at healing bone defects. There has been a remarkable advancement in producing bone substitutes in the tissue engineering area, but the sufficient and timely vascularization of engineered constructs for optimal tissue integration and regeneration is lacking due to mismatch in the scaffold characteristics and new bone tissue reconstruction. Therefore, various key challenges remain to be overcome. A deep understanding of angiogenesis and osteogenesis progressions is required to manufacture bone hydrogels with satisfactory results. The current review briefly discusses the fundamentals of bone tissues, the significance of angiogenesis-osteogenesis progressions and their inducers, the efficacy of biomaterials and composite hydrogel-promoted neo-vasculogenesis (i.e. angiogenesis) and bone mineralization (i.e. osteogenesis), and related challenges, including future research directions.
Collapse
Affiliation(s)
- Anuj Kumar
- School of Chemical Engineering, Yeungnam University, 280 Daehak-ro, Gyeongsan 38541, South Korea. .,Research Institute of Cell Culture, Yeungnam University, 280 Daehak-ro, Gyeongsan 38541, South Korea
| | - Ankur Sood
- School of Chemical Engineering, Yeungnam University, 280 Daehak-ro, Gyeongsan 38541, South Korea.
| | - Ritu Singhmar
- School of Chemical Engineering, Yeungnam University, 280 Daehak-ro, Gyeongsan 38541, South Korea.
| | - Yogendra Kumar Mishra
- Smart Materials, NanoSYD, Mads Clausen Institute, University of Southern Denmark, Alsion 2, 6400, Sønderborg, Denmark
| | - Vijay Kumar Thakur
- Biorefining and Advanced Materials Research Center, Scotland's Rural College (SRUC), Kings Buildings, Edinburgh EH9 3JG, UK.,School of Engineering, University of Petroleum and Energy Studies (UPES), Dehradun 248007, Uttarakhand, India
| | - Sung Soo Han
- School of Chemical Engineering, Yeungnam University, 280 Daehak-ro, Gyeongsan 38541, South Korea. .,Research Institute of Cell Culture, Yeungnam University, 280 Daehak-ro, Gyeongsan 38541, South Korea
| |
Collapse
|
15
|
Cui J, Yu X, Yu B, Yang X, Fu Z, Wan J, Zhu M, Wang X, Lin K. Coaxially Fabricated Dual-Drug Loading Electrospinning Fibrous Mat with Programmed Releasing Behavior to Boost Vascularized Bone Regeneration. Adv Healthc Mater 2022; 11:e2200571. [PMID: 35668705 DOI: 10.1002/adhm.202200571] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 05/22/2022] [Indexed: 01/24/2023]
Abstract
In clinical treatment, the bone regeneration of critical-size defects is desiderated to be solved, and the regeneration of large bone segment defects depends on early vascularization. Therefore, overcoming insufficient vascularization in artificial bone grafts may be a promising strategy for critical-size bone regeneration. Herein, a novel dual-drug programmed releasing electrospinning fibrous mat (EFM) with a deferoxamine (DFO)-loaded shell layer and a dexamethasone (DEX)-loaded core layer is fabricated using coaxial electrospinning technology, considering the temporal sequence of vascularization and bone repair. DFO acts as an angiogenesis promoter and DEX is used as an osteogenesis inducer. The results demonstrate that the early and rapid release of DFO promotes angiogenesis in human umbilical vascular endothelial cells and the sustained release of DEX enhances the osteogenic differentiation of rat bone mesenchymal stem cells. DFO and DEX exert synergetic effects on osteogenic differentiation via the Wnt/β-catenin signaling pathway, and the dual-drug programmed releasing EFM acquired perfect vascularized bone regeneration ability in a rat calvarial defect model. Overall, the study suggests a low-cost strategy to enhance vascularized bone regeneration by adjusting the behavior of angiogenesis and osteogenesis in time dimension.
Collapse
Affiliation(s)
- Jinjie Cui
- Department of Oral and Cranio-Maxillofacial Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, Shanghai, 200011, China
| | - Xingge Yu
- Department of Oral and Cranio-Maxillofacial Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, Shanghai, 200011, China
| | - Bin Yu
- Department of Oral and Cranio-Maxillofacial Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, Shanghai, 200011, China
| | - Xiuyi Yang
- Department of Orthodontics, Affiliated Stomatological Hospital of Soochow University, Suzhou, 215005, China
| | - Zeyu Fu
- Department of Oral and Cranio-Maxillofacial Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, Shanghai, 200011, China
| | - Jianyu Wan
- Department of Oral and Cranio-Maxillofacial Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, Shanghai, 200011, China
| | - Min Zhu
- Department of Oral and Cranio-Maxillofacial Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, Shanghai, 200011, China
| | - Xudong Wang
- Department of Oral and Cranio-Maxillofacial Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, Shanghai, 200011, China
| | - Kaili Lin
- Department of Oral and Cranio-Maxillofacial Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, Shanghai, 200011, China
| |
Collapse
|
16
|
Self-Assembled Peptide Nanostructures for ECM Biomimicry. NANOMATERIALS 2022; 12:nano12132147. [PMID: 35807982 PMCID: PMC9268130 DOI: 10.3390/nano12132147] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Revised: 06/18/2022] [Accepted: 06/21/2022] [Indexed: 02/04/2023]
Abstract
Proteins are functional building blocks of living organisms that exert a wide variety of functions, but their synthesis and industrial production can be cumbersome and expensive. By contrast, short peptides are very convenient to prepare at a low cost on a large scale, and their self-assembly into nanostructures and gels is a popular avenue for protein biomimicry. In this Review, we will analyze the last 5-year progress on the incorporation of bioactive motifs into self-assembling peptides to mimic functional proteins of the extracellular matrix (ECM) and guide cell fate inside hydrogel scaffolds.
Collapse
|
17
|
Piras CC, Mahon CS, Genever PG, Smith DK. Shaping and Patterning Supramolecular Materials─Stem Cell-Compatible Dual-Network Hybrid Gels Loaded with Silver Nanoparticles. ACS Biomater Sci Eng 2022; 8:1829-1840. [PMID: 35364810 PMCID: PMC9092345 DOI: 10.1021/acsbiomaterials.1c01560] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
![]()
Hydrogels
with spatio-temporally
controlled properties are appealing
materials for biological and pharmaceutical applications. We make
use of mild acidification protocols to fabricate hybrid gels using
calcium alginate in the presence of a preformed thermally triggered
gel based on a low-molecular-weight gelator (LMWG) 1,3:2:4-di(4-acylhydrazide)-benzylidene
sorbitol (DBS-CONHNH2). Nonwater-soluble calcium carbonate
slowly releases calcium ions over time when exposed to an acidic pH,
triggering the assembly of the calcium alginate gel network. We combined
the gelators in different ways: (i) the LMWG was used as a template
to spatially control slow calcium alginate gelation within preformed
gel beads, using glucono-δ-lactone (GdL) to lower the pH; (ii)
the LMWG was used as a template to spatially control slow calcium
alginate gelation within preformed gel trays, using diphenyliodonium
nitrate (DPIN) as a photoacid to lower the pH, and spatial resolution
was achieved by masking. The dual-network hybrid gels display highly
tunable properties, and the beads are compatible with stem cell growth.
Furthermore, they preserve the LMWG function of inducing in situ silver
nanoparticle (AgNP) formation, which provides the gels with antibacterial
activity. These gels have potential for eventual regenerative medicine
applications in (e.g.) bone tissue engineering.
Collapse
Affiliation(s)
- Carmen C Piras
- Department of Chemistry, University of York, Heslington, York YO10 5DD, United Kingdom
| | - Clare S Mahon
- Department of Chemistry, University of York, Heslington, York YO10 5DD, United Kingdom
| | - Paul G Genever
- Department of Biology, University of York, Heslington, York YO10 5DD, United Kingdom
| | - David K Smith
- Department of Chemistry, University of York, Heslington, York YO10 5DD, United Kingdom
| |
Collapse
|
18
|
Hao Z, Li H, Wang Y, Hu Y, Chen T, Zhang S, Guo X, Cai L, Li J. Supramolecular Peptide Nanofiber Hydrogels for Bone Tissue Engineering: From Multihierarchical Fabrications to Comprehensive Applications. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2103820. [PMID: 35128831 PMCID: PMC9008438 DOI: 10.1002/advs.202103820] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 01/02/2022] [Indexed: 05/03/2023]
Abstract
Bone tissue engineering is becoming an ideal strategy to replace autologous bone grafts for surgical bone repair, but the multihierarchical complexity of natural bone is still difficult to emulate due to the lack of suitable biomaterials. Supramolecular peptide nanofiber hydrogels (SPNHs) are emerging biomaterials because of their inherent biocompatibility, satisfied biodegradability, high purity, facile functionalization, and tunable mechanical properties. This review initially focuses on the multihierarchical fabrications by SPNHs to emulate natural bony extracellular matrix. Structurally, supramolecular peptides based on distinctive building blocks can assemble into nanofiber hydrogels, which can be used as nanomorphology-mimetic scaffolds for tissue engineering. Biochemically, bioactive motifs and bioactive factors can be covalently tethered or physically absorbed to SPNHs to endow various functions depending on physiological and pharmacological requirements. Mechanically, four strategies are summarized to optimize the biophysical microenvironment of SPNHs for bone regeneration. Furthermore, comprehensive applications about SPNHs for bone tissue engineering are reviewed. The biomaterials can be directly used in the form of injectable hydrogels or composite nanoscaffolds, or they can be used to construct engineered bone grafts by bioprinting or bioreactors. Finally, continuing challenges and outlook are discussed.
Collapse
Affiliation(s)
- Zhuowen Hao
- Department of OrthopedicsZhongnan Hospital of Wuhan UniversityDonghu Road 169Wuhan430071China
| | - Hanke Li
- Department of OrthopedicsZhongnan Hospital of Wuhan UniversityDonghu Road 169Wuhan430071China
| | - Yi Wang
- Department of OrthopedicsZhongnan Hospital of Wuhan UniversityDonghu Road 169Wuhan430071China
| | - Yingkun Hu
- Department of OrthopedicsZhongnan Hospital of Wuhan UniversityDonghu Road 169Wuhan430071China
| | - Tianhong Chen
- Department of OrthopedicsZhongnan Hospital of Wuhan UniversityDonghu Road 169Wuhan430071China
| | - Shuwei Zhang
- Department of OrthopedicsZhongnan Hospital of Wuhan UniversityDonghu Road 169Wuhan430071China
| | - Xiaodong Guo
- Department of OrthopedicsUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyJiefang Road 1277Wuhan430022China
| | - Lin Cai
- Department of OrthopedicsZhongnan Hospital of Wuhan UniversityDonghu Road 169Wuhan430071China
| | - Jingfeng Li
- Department of OrthopedicsZhongnan Hospital of Wuhan UniversityDonghu Road 169Wuhan430071China
| |
Collapse
|
19
|
Multicomponent Interpenetrating metal based Alginate-Carrageenan biopolymer Hydrogel beads substantiated by Graphene oxide for efficient removal of Methylene Blue from waste water. Chem Eng Res Des 2022. [DOI: 10.1016/j.cherd.2022.04.017] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
20
|
Bu W, Wu Y, Ghaemmaghami AM, Sun H, Mata A. Rational design of hydrogels for immunomodulation. Regen Biomater 2022; 9:rbac009. [PMID: 35668923 PMCID: PMC9160883 DOI: 10.1093/rb/rbac009] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 01/21/2022] [Accepted: 01/30/2022] [Indexed: 11/13/2022] Open
Abstract
Abstract
The immune system protects organisms against endogenous and exogenous harm and plays a key role in tissue development, repair, and regeneration. Traditional immunomodulatory biologics exhibit limitations including degradation by enzymes, short half-life, and lack of targeting ability. Encapsulating or binding these biologics within biomaterials is an effective way to address these problems. Hydrogels are promising immunomodulatory materials because of their prominent biocompatibility, tuneability, and versatility. However, to take advantage of these opportunities and optimize material performance, it is important to more specifically elucidate, and leverage on, how hydrogels affect and control the immune response. Here, we summarize how key physical and chemical properties of hydrogels affect the immune response. We first provide an overview of underlying steps of the host immune response upon exposure to biomaterials. Then, we discuss recent advances in immunomodulatory strategies where hydrogels play a key role through a) physical properties including dimensionality, stiffness, porosity, and topography; b) chemical properties including wettability, electric property, and molecular presentation; and c) the delivery of bioactive molecules via chemical or physical cues. Thus, this review aims to build a conceptual and practical toolkit for the design of immune-instructive hydrogels capable of modulating the host immune response.
Collapse
Affiliation(s)
- Wenhuan Bu
- Liaoning Provincial Key Laboratory of Oral Diseases, School of Stomatology, China Medical University, Shenyang, 110001, China
- Department of Dental Materials, School of Stomatology, China Medical University, Shenyang, 110001, China
- Department of Center Laboratory, School of Stomatology, China Medical University, Shenyang, 110001, China
- School of Pharmacy, University of Nottingham, Nottingham, NG7 2RD, UK
- Biodiscovery Institute, University of Nottingham, Nottingham, NG7 2RD, UK
| | - Yuanhao Wu
- School of Pharmacy, University of Nottingham, Nottingham, NG7 2RD, UK
- Biodiscovery Institute, University of Nottingham, Nottingham, NG7 2RD, UK
| | - Amir M Ghaemmaghami
- Division of Immunology, School of Life Sciences, University of Nottingham, Nottingham, NG7 2RD, UK
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA, 90024, USA
| | - Hongchen Sun
- Liaoning Provincial Key Laboratory of Oral Diseases, School of Stomatology, China Medical University, Shenyang, 110001, China
| | - Alvaro Mata
- School of Pharmacy, University of Nottingham, Nottingham, NG7 2RD, UK
- Biodiscovery Institute, University of Nottingham, Nottingham, NG7 2RD, UK
- Department of Chemical and Environmental Engineering, University of Nottingham, Nottingham, NG7 2RD, UK
| |
Collapse
|
21
|
Leistner AL, Kistner DG, Fengler C, Pianowski ZL. Reversible photodissipation of composite photochromic azobenzene-alginate supramolecular hydrogels. RSC Adv 2022; 12:4771-4776. [PMID: 35425487 PMCID: PMC8981262 DOI: 10.1039/d1ra09218a] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 02/01/2022] [Indexed: 01/01/2023] Open
Abstract
Supramolecular smart materials can quickly elicit macroscopic changes upon external stimulation. Here we report that an azobenzene-containing cyclic dipeptide can form composite supramolecular hydrogels with alginate based on the charge complementarity, at lower loading than the critical gelation concentrations of either component. The gels can reversibly dissipate to fluids with UV light. They can also encapsulate and photorelease fluorescent cargo. Upon treatment of the gels with aqueous calcium salts, the alginate component is permanently cross-linked and the photochromic component is solubilized.
Collapse
Affiliation(s)
- Anna-Lena Leistner
- Institut für Organische Chemie Karlsruher Institut für Technologie Campus Süd, Fritz-Haber-Weg 6 76131 Karlsruhe Germany
| | - David Georg Kistner
- Institut für Organische Chemie Karlsruher Institut für Technologie Campus Süd, Fritz-Haber-Weg 6 76131 Karlsruhe Germany
| | - Christian Fengler
- Institut für Technische Chemie and Polymerchemie Karlsruher Institut für Technologie Campus Süd, Engesserstraße 18 76128 Karlsruhe Germany
| | - Zbigniew L Pianowski
- Institut für Organische Chemie Karlsruher Institut für Technologie Campus Süd, Fritz-Haber-Weg 6 76131 Karlsruhe Germany
- Institute of Biological and Chemical Systems - FMS Karlsruher Institut für Technologie Campus Nord, Hermann-von-Helmholtz Platz 1, 76344 Eggenstein-Leopoldshafen Germany
| |
Collapse
|
22
|
Redondo-Gómez C, Padilla-Lopátegui S, Mata A, Azevedo HS. Peptide Amphiphile Hydrogels Based on Homoternary Cucurbit[8]uril Host-Guest Complexes. Bioconjug Chem 2022; 33:111-120. [PMID: 34914370 DOI: 10.1021/acs.bioconjchem.1c00441] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Supramolecular hydrogels based on peptide amphiphiles (PAs) are promising materials for tissue engineering and model extracellular matrixes for biological studies. While PA hydrogels are conventionally formed via electrostatic screening, new hydrogelation mechanisms might help to improve the design and functionality of these materials. Here, we present a host-guest-mediated PA hydrogelation method that relies on the formation of a host-guest homoternary complex with cucurbit[8]uril (CB[8]) and aromatic amino-acid-bearing PA nanofibers. As a result of the host-guest cross-linking between PA nanofibers, hierarchical morphologies and increased stiffness were found when host-guest-mediated PA hydrogels were compared to their ion-based equivalents. Additionally, both families of hydrogels exhibited similar biocompatibilities. These results demonstrate that CB[8]-mediated hydrogelation can be used as an alternative cross-linking method to upgrade the design of PA materials and extend their biomedical applications.
Collapse
Affiliation(s)
- Carlos Redondo-Gómez
- School of Engineering and Materials Science, Queen Mary University of London, London E1 4NS, U.K
- Institute of Bioengineering, Queen Mary University of London, London E1 4NS, U.K
- National Nanotechnology Laboratory LANOTEC, National Center for High Technology CeNAT, 1174-1200 Pavas, San José 10109, Costa Rica
| | - Soraya Padilla-Lopátegui
- School of Engineering and Materials Science, Queen Mary University of London, London E1 4NS, U.K
- Institute of Bioengineering, Queen Mary University of London, London E1 4NS, U.K
| | - Alvaro Mata
- School of Engineering and Materials Science, Queen Mary University of London, London E1 4NS, U.K
- Institute of Bioengineering, Queen Mary University of London, London E1 4NS, U.K
- School of Pharmacy, University of Nottingham, University Park, Nottingham NG7 2RD, U.K
- Department of Chemical and Environmental Engineering, University of Nottingham, University Park, Nottingham NG7 2RD, U.K
- Biodiscovery Institute, University of Nottingham, University Park, Nottingham NG7 2RD, U.K
| | - Helena S Azevedo
- School of Engineering and Materials Science, Queen Mary University of London, London E1 4NS, U.K
- Institute of Bioengineering, Queen Mary University of London, London E1 4NS, U.K
| |
Collapse
|
23
|
Ghosh M, Majkowska A, Mirsa R, Bera S, Rodríguez-Cabello JC, Mata A, Adler-Abramovich L. Disordered Protein Stabilization by Co-Assembly of Short Peptides Enables Formation of Robust Membranes. ACS APPLIED MATERIALS & INTERFACES 2022; 14:464-473. [PMID: 34941264 DOI: 10.1021/acsami.1c22136] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Molecular self-assembly is a spontaneous natural process resulting in highly ordered nano to microarchitectures. We report temperature-independent formation of robust stable membranes obtained by the spontaneous interaction of intrinsically disordered elastin-like polypeptides (ELPs) with short aromatic peptides at temperatures both below and above the conformational transition temperature of the ELPs. The membranes are stable over time and display durability over a wide range of parameters including temperature, pH, and ultrasound energy. The morphology and composition of the membranes were analyzed using microscopy. These robust structures support preosteoblast cell adhesion and proliferation as well as pH-dependent cargo release. Simple noncovalent interactions with short aromatic peptides can overcome conformational restrictions due to the phase transition to facilitate the formation of complex bioactive scaffolds that are stable over a wide range of environmental parameters. This approach offers novel possibilities for controlling the conformational restriction of intrinsically disordered proteins and using them in the design of new materials.
Collapse
Affiliation(s)
- Moumita Ghosh
- Department of Oral Biology, The Goldschleger School of Dental Medicine, Sackler Faculty of Medicine, Tel-Aviv University, Tel Aviv 69978, Israel
- The Centre for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv 6997801, Israel
- Department of Chemistry, Techno India University, EM-4, EM Block, Sector V, Bidhannagar, Kolkata, West Bengal 700091, India
| | - Anna Majkowska
- School of Engineering & Materials Science, Queen Mary University of London, London E1 4NS, U.K
- William Harvey Research Institute, Queen Mary University of London, London EC1M 6BQ, U.K
| | - Rajkumar Mirsa
- Department of Oral Biology, The Goldschleger School of Dental Medicine, Sackler Faculty of Medicine, Tel-Aviv University, Tel Aviv 69978, Israel
- The Centre for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Santu Bera
- Department of Oral Biology, The Goldschleger School of Dental Medicine, Sackler Faculty of Medicine, Tel-Aviv University, Tel Aviv 69978, Israel
- The Centre for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv 6997801, Israel
| | | | - Alvaro Mata
- School of Pharmacy, University of Nottingham, Nottingham NG7 2RD, U.K
- Department of Chemical and Environmental Engineering, University of Nottingham, Nottingham NG7 2RD, U.K
- Biodiscovery Institute, University of Nottingham, Nottingham NG7 2RD, U.K
| | - Lihi Adler-Abramovich
- Department of Oral Biology, The Goldschleger School of Dental Medicine, Sackler Faculty of Medicine, Tel-Aviv University, Tel Aviv 69978, Israel
- The Centre for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv 6997801, Israel
| |
Collapse
|
24
|
Soft-Tissue-Mimicking Using Hydrogels for the Development of Phantoms. Gels 2022; 8:gels8010040. [PMID: 35049575 PMCID: PMC8774477 DOI: 10.3390/gels8010040] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2021] [Revised: 12/20/2021] [Accepted: 01/01/2022] [Indexed: 12/11/2022] Open
Abstract
With the currently available materials and technologies it is difficult to mimic the mechanical properties of soft living tissues. Additionally, another significant problem is the lack of information about the mechanical properties of these tissues. Alternatively, the use of phantoms offers a promising solution to simulate biological bodies. For this reason, to advance in the state-of-the-art a wide range of organs (e.g., liver, heart, kidney as well as brain) and hydrogels (e.g., agarose, polyvinyl alcohol –PVA–, Phytagel –PHY– and methacrylate gelatine –GelMA–) were tested regarding their mechanical properties. For that, viscoelastic behavior, hardness, as well as a non-linear elastic mechanical response were measured. It was seen that there was a significant difference among the results for the different mentioned soft tissues. Some of them appear to be more elastic than viscous as well as being softer or harder. With all this information in mind, a correlation between the mechanical properties of the organs and the different materials was performed. The next conclusions were drawn: (1) to mimic the liver, the best material is 1% wt agarose; (2) to mimic the heart, the best material is 2% wt agarose; (3) to mimic the kidney, the best material is 4% wt GelMA; and (4) to mimic the brain, the best materials are 4% wt GelMA and 1% wt agarose. Neither PVA nor PHY was selected to mimic any of the studied tissues.
Collapse
|
25
|
Sari B, Isik M, Eylem CC, Kilic C, Okesola BO, Karakaya E, Emregul E, Nemutlu E, Derkus B. Omics Technologies for High-Throughput-Screening of Cell-Biomaterial Interactions. Mol Omics 2022; 18:591-615. [DOI: 10.1039/d2mo00060a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Recent research effort in biomaterial development has largely focused on engineering bio-instructive materials to stimulate specific cell signaling. Assessing the biological performance of these materials using time-consuming and trial-and-error traditional...
Collapse
|
26
|
Isik M, Eylem CC, Haciefendioglu T, Yildirim E, Sari B, Nemutlu E, Emregul E, Okesola BO, Derkus B. Mechanically robust hybrid hydrogels of photo-crosslinkable gelatin and laminin-mimetic peptide amphiphiles for neural induction. Biomater Sci 2021; 9:8270-8284. [PMID: 34766605 DOI: 10.1039/d1bm01350e] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Self-assembling bio-instructive materials that can provide a biomimetic tissue microenvironment with the capability to regulate cellular behaviors represent an attractive platform in regenerative medicine. Herein, we develop a hybrid neuro-instructive hydrogel that combines the properties of a photo-crosslinkable gelatin methacrylate (GelMA) and self-assembling peptide amphiphiles (PAs) bearing a laminin-derived neuro-inductive epitope (PA-GSR). Electrostatic interaction and ultraviolet light crosslinking mechanisms were combined to create dual-crosslinked hybrid hydrogels with tunable stiffness. Spectroscopic, microscopic and theoretical techniques show that the cationic PA-GSR(+) electrostatically co-assembles with the negatively charged GelMA to create weak hydrogels with hierarchically ordered microstructures, which were further photo-crosslinked to create mechanically robust hydrogels. Dynamic oscillatory rheology and micromechanical testing show that photo-crosslinking of the co-assembled GelMA and PA-GSR(+) hydrogel results in robust hydrogels displaying improved stiffness. Gene expression analysis was used to show that GelMA/PA-GSR(+) hydrogels can induce human mesenchymal stem cells (hMSCs) into neural-lineage cells and supports neural-lineage specification of neuroblast-like cells (SH-SY5Y) in a growth-factor-free manner. Also, metabolomics analysis suggests that the hydrogel alters the metabolite profiles in the cells by affecting multiple molecular pathways. This work highlights a new approach for the design of PA-based hybrid hydrogels with robust mechanical properties and biological functionalities for nerve tissue regeneration.
Collapse
Affiliation(s)
- Melis Isik
- Department of Chemistry, Faculty of Science, Ankara University, 06560 Ankara, Turkey.
| | - Cemil Can Eylem
- Analytical Chemistry Division, Faculty of Pharmacy, Hacettepe University, 06230 Ankara, Turkey
| | | | - Erol Yildirim
- Chemistry Department, Middle East Technical University, 06800 Ankara, Turkey.,Department of Polymer Science and Technology, Middle East Technical University, 06800 Ankara, Turkey.,Department of Micro and Nanotechnology, Middle East Technical University, 06800 Ankara, Turkey
| | - Buse Sari
- Department of Chemistry, Faculty of Science, Ankara University, 06560 Ankara, Turkey. .,Stem Cell Research Lab, Department of Chemistry, Faculty of Science, Ankara University, 06560 Ankara, Turkey
| | - Emirhan Nemutlu
- Analytical Chemistry Division, Faculty of Pharmacy, Hacettepe University, 06230 Ankara, Turkey.,Bioanalytic and Omics Laboratory, Faculty of Pharmacy, Hacettepe University, Ankara, Turkey
| | - Emel Emregul
- Department of Chemistry, Faculty of Science, Ankara University, 06560 Ankara, Turkey.
| | - Babatunde O Okesola
- Institute of Bioengineering, Queen Mary University of London, London, E1 4NS, UK. .,School of Engineering and Materials Science, Queen Mary University of London, London, E1 4NS, UK
| | - Burak Derkus
- Department of Chemistry, Faculty of Science, Ankara University, 06560 Ankara, Turkey. .,Stem Cell Research Lab, Department of Chemistry, Faculty of Science, Ankara University, 06560 Ankara, Turkey
| |
Collapse
|
27
|
Stahl A, Yang YP. Regenerative Approaches for the Treatment of Large Bone Defects. TISSUE ENGINEERING. PART B, REVIEWS 2021; 27:539-547. [PMID: 33138705 PMCID: PMC8739850 DOI: 10.1089/ten.teb.2020.0281] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Accepted: 11/02/2020] [Indexed: 12/15/2022]
Abstract
A variety of engineered materials have gained acceptance in orthopedic practice as substitutes for autologous bone grafts, although the regenerative efficacy of these engineered grafts is still limited compared with that of transplanted native tissues. For bone defects greater than 4-5 cm, however, common bone grafting procedures are insufficient and more complicated surgical interventions are required to repair and regenerate the damaged or missing bone. In this review, we describe current grafting materials and surgical techniques for the reconstruction of large bone defects, followed by tissue engineering (TE) efforts to develop improved therapies. Particular emphasis is placed on graft vascularization, because for both autologous bone and engineered alternatives, achieving adequate vascular development within the regenerating bone tissues remains a significant challenge in the context of large bone defects. To this end, TE and surgical strategies to induce development of a vasculature within bone grafts are discussed. Impact statement This review aims to present an accessible and thorough overview of current orthopedic surgical techniques as well as bone tissue engineering and vascularization strategies that might one day offer improvements to clinical therapies for the repair of large bone defects. We consider the lessons that clinical orthopedic reconstructive practices can contribute to the push toward engineered bone.
Collapse
Affiliation(s)
- Alexander Stahl
- Department of Orthopaedic Surgery, Stanford University, Stanford, California, USA
- Department of Chemistry, Stanford University, Stanford, California, USA
| | - Yunzhi Peter Yang
- Department of Orthopaedic Surgery, Stanford University, Stanford, California, USA
- Department of Materials Science and Engineering, and Stanford University, Stanford, California, USA
- Department of Bioengineering, Stanford University, Stanford, California, USA
| |
Collapse
|
28
|
Walsh CM, Wychowaniec JK, Brougham DF, Dooley D. Functional hydrogels as therapeutic tools for spinal cord injury: New perspectives on immunopharmacological interventions. Pharmacol Ther 2021; 234:108043. [PMID: 34813862 DOI: 10.1016/j.pharmthera.2021.108043] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 11/10/2021] [Accepted: 11/11/2021] [Indexed: 02/06/2023]
Abstract
Spinal cord injury (SCI) is a complex medical and psychological challenge for which there is no curative therapy currently available. Despite major progress in pharmacological and surgical approaches, clinical trials for SCI patients have been uniformly disappointing thus far as there are many practical and biological issues yet to be resolved. Neuroinflammation is a critical event of the secondary injury phase after SCI, and recent research strategies have focused on modulating the immune response after injury to provide a more favorable recovery environment. Biomaterials can serve this purpose by providing physical and trophic support to the injured spinal cord after SCI. Of all potential biomaterials, functional hydrogels are emerging as a key component in novel treatment strategies for SCI, including controlled and localized delivery of immunomodulatory therapies to drive polarization of immune cells towards a pro-regenerative phenotype. Here, we extensively review recent developments in the use of functional hydrogels as immunomodulatory therapies for SCI. We briefly describe physicochemical properties of hydrogels and demonstrate how advanced fabrication methods lead to the required heterogeneity and hierarchical arrangements that increasingly mimic complex spinal cord tissue. We then summarize potential SCI therapeutic modalities including: (i) hydrogels alone; (ii) hydrogels as cellular or (iii) bioactive molecule delivery vehicles, and; (iv) combinatorial approaches. By linking the structural properties of hydrogels to their functions in treatment with particular focus on immunopharmacological stimuli, this may accelerate further development of functional hydrogels for SCI, and indeed next-generation central nervous system regenerative therapies.
Collapse
Affiliation(s)
- Ciara M Walsh
- School of Medicine, Health Sciences Centre, University College Dublin, Belfield, Dublin 4, Ireland; UCD Conway Institute of Biomolecular & Biomedical Research, University College Dublin, Belfield, Dublin 4, Ireland
| | - Jacek K Wychowaniec
- School of Chemistry, University College Dublin, Belfield, Dublin 4, Ireland; AO Research Institute Davos, Clavadelerstrasse 8, 7270 Davos, Switzerland
| | - Dermot F Brougham
- School of Chemistry, University College Dublin, Belfield, Dublin 4, Ireland
| | - Dearbhaile Dooley
- School of Medicine, Health Sciences Centre, University College Dublin, Belfield, Dublin 4, Ireland; UCD Conway Institute of Biomolecular & Biomedical Research, University College Dublin, Belfield, Dublin 4, Ireland.
| |
Collapse
|
29
|
Mendoza-Martinez AK, Loessner D, Mata A, Azevedo HS. Modeling the Tumor Microenvironment of Ovarian Cancer: The Application of Self-Assembling Biomaterials. Cancers (Basel) 2021; 13:5745. [PMID: 34830897 PMCID: PMC8616551 DOI: 10.3390/cancers13225745] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 11/07/2021] [Accepted: 11/11/2021] [Indexed: 02/06/2023] Open
Abstract
Ovarian cancer (OvCa) is one of the leading causes of gynecologic malignancies. Despite treatment with surgery and chemotherapy, OvCa disseminates and recurs frequently, reducing the survival rate for patients. There is an urgent need to develop more effective treatment options for women diagnosed with OvCa. The tumor microenvironment (TME) is a key driver of disease progression, metastasis and resistance to treatment. For this reason, 3D models have been designed to represent this specific niche and allow more realistic cell behaviors compared to conventional 2D approaches. In particular, self-assembling peptides represent a promising biomaterial platform to study tumor biology. They form nanofiber networks that resemble the architecture of the extracellular matrix and can be designed to display mechanical properties and biochemical motifs representative of the TME. In this review, we highlight the properties and benefits of emerging 3D platforms used to model the ovarian TME. We also outline the challenges associated with using these 3D systems and provide suggestions for future studies and developments. We conclude that our understanding of OvCa and advances in materials science will progress the engineering of novel 3D approaches, which will enable the development of more effective therapies.
Collapse
Affiliation(s)
- Ana Karen Mendoza-Martinez
- School of Engineering and Materials Science, Queen Mary University of London, Mile End Road, London E1 4NS, UK;
- Institute of Bioengineering, Queen Mary University of London, Mile End Road, London E1 4NS, UK
| | - Daniela Loessner
- Department of Chemical Engineering, Faculty of Engineering, Monash University, Melbourne, VIC 3800, Australia;
- Department of Materials Science and Engineering, Faculty of Engineering, Monash University, Melbourne, VIC 3800, Australia
- Department of Anatomy and Developmental Biology, Biomedicine Discovery Institute, Faculty of Medicine, Nursing and Health Sciences, Monash University, Melbourne, VIC 3800, Australia
- Max Bergmann Center of Biomaterials Dresden, Leibniz Institute of Polymer Research Dresden e.V., 01069 Dresden, Germany
| | - Alvaro Mata
- School of Pharmacy, University of Nottingham, Nottingham NG7 2RD, UK;
- Department of Chemical and Environmental Engineering, University of Nottingham, Nottingham NG7 2RD, UK
- Biodiscovery Institute, University of Nottingham, Nottingham NG7 2RD, UK
| | - Helena S. Azevedo
- School of Engineering and Materials Science, Queen Mary University of London, Mile End Road, London E1 4NS, UK;
- Institute of Bioengineering, Queen Mary University of London, Mile End Road, London E1 4NS, UK
| |
Collapse
|
30
|
Exploiting the fundamentals of biological organization for the advancement of biofabrication. Curr Opin Biotechnol 2021; 74:42-54. [PMID: 34798447 DOI: 10.1016/j.copbio.2021.10.016] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 09/26/2021] [Accepted: 10/14/2021] [Indexed: 12/12/2022]
Abstract
The field of biofabrication continues to progress, offering higher levels of spatial control, reproducibility, and functionality. However, we remain far from recapitulating what nature has achieved. Biological systems such as tissues and organs are assembled from the bottom-up through coordinated supramolecular and cellular processes that result in their remarkable structures and functionalities. In this perspective, we propose that incorporating such biological assembling mechanisms within fabrication techniques, offers an opportunity to push the boundaries of biofabrication. We dissect these mechanisms into distinct biological organization principles (BOPs) including self-assembly, compartmentalization, diffusion-reaction, disorder-to-order transitions, and out-of-equilibrium processes. We highlight recent work demonstrating the viability and potential of these approaches to enhance scalability, reproducibility, vascularization, and biomimicry; as well as current challenges to overcome.
Collapse
|
31
|
Dodda JM, Azar MG, Sadiku R. Crosslinking Trends in Multicomponent Hydrogels for Biomedical Applications. Macromol Biosci 2021; 21:e2100232. [PMID: 34612608 DOI: 10.1002/mabi.202100232] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 09/09/2021] [Indexed: 12/15/2022]
Abstract
Multicomponent-based hydrogels are well established candidates for biomedical applications. However, certain aspects of multicomponent systems, e.g., crosslinking, structural binding, network formation, proteins/drug incorporation, etc., are challenging aspects to modern biomedical research. The types of crosslinking and network formation are crucial for the effective combination of multiple component systems. The creation of a complex system in the overall structure and the crosslinking efficiency of different polymeric chains in an organized fashion are crucially important, especially when the materials are for biomedical applications. Therefore, the engineering of hydrogel has to be, succinctly understood, carefully formulated, and expertly designed. The different crosslinking methods in use, hydrogen bonding, electrostatic interaction, coordination bonding, and self-assembly. The formations of double, triple, and multiple networks, are well established. A systematic study of the crosslinking mechanisms in multicomponent systems, in terms of the crosslinking types, network formation, intramolecular bonds between different structural units, and their potentials for biomedical applications, is lacking and therefore, these aspects require investigations. To this end, the present review, focuses on the recent advances in areas of the physical, chemical, and enzymatic crosslinking methods that are often, employed for the designing of multicomponent hydrogels.
Collapse
Affiliation(s)
- Jagan Mohan Dodda
- New Technologies-Research Centre (NTC), University of West Bohemia, Univerzitní 8, Pilsen, 301 00, Czech Republic
| | - Mina Ghafouri Azar
- New Technologies-Research Centre (NTC), University of West Bohemia, Univerzitní 8, Pilsen, 301 00, Czech Republic
| | - Rotimi Sadiku
- Institute of NanoEngineering Research (INER) and Department of Chemical, Metallurgical and Materials Engineering, Tshwane University of Technology, Staatsartillerie Rd, Pretoria West Campus, Pretoria, 0183, Republic of South Africa
| |
Collapse
|
32
|
Ajovalasit A, Redondo-Gómez C, Sabatino MA, Okesola BO, Braun K, Mata A, Dispenza C. Carboxylated-xyloglucan and peptide amphiphile co-assembly in wound healing. Regen Biomater 2021; 8:rbab040. [PMID: 34386265 PMCID: PMC8355605 DOI: 10.1093/rb/rbab040] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 06/14/2021] [Accepted: 06/23/2021] [Indexed: 12/13/2022] Open
Abstract
Hydrogel wound dressings can play critical roles in wound healing protecting the wound from trauma or contamination and providing an ideal environment to support the growth of endogenous cells and promote wound closure. This work presents a self-assembling hydrogel dressing that can assist the wound repair process mimicking the hierarchical structure of skin extracellular matrix. To this aim, the co-assembly behaviour of a carboxylated variant of xyloglucan (CXG) with a peptide amphiphile (PA-H3) has been investigated to generate hierarchical constructs with tuneable molecular composition, structure, and properties. Transmission electron microscopy and circular dichroism at a low concentration shows that CXG and PA-H3 co-assemble into nanofibres by hydrophobic and electrostatic interactions and further aggregate into nanofibre bundles and networks. At a higher concentration, CXG and PA-H3 yield hydrogels that have been characterized for their morphology by scanning electron microscopy and for the mechanical properties by small-amplitude oscillatory shear rheological measurements and compression tests at different CXG/PA-H3 ratios. A preliminary biological evaluation has been carried out both in vitro with HaCat cells and in vivo in a mouse model.
Collapse
Affiliation(s)
- Alessia Ajovalasit
- Dipartimento di Ingegneria (DI), Università degli Studi di Palermo, Viale delle Scienze, Edificio 6, Palermo 90128, Italy
- School of Engineering & Materials Science, Queen Mary University of London, London E1 4NS, UK
- Institute of Bioengineering, Queen Mary University of London, London E1 4NS, UK
| | - Carlos Redondo-Gómez
- School of Engineering & Materials Science, Queen Mary University of London, London E1 4NS, UK
- Institute of Bioengineering, Queen Mary University of London, London E1 4NS, UK
| | - Maria Antonietta Sabatino
- Dipartimento di Ingegneria (DI), Università degli Studi di Palermo, Viale delle Scienze, Edificio 6, Palermo 90128, Italy
| | - Babatunde O Okesola
- School of Engineering & Materials Science, Queen Mary University of London, London E1 4NS, UK
- Institute of Bioengineering, Queen Mary University of London, London E1 4NS, UK
| | - Kristin Braun
- Blizard Institute, Barts and The London School of Medicine and Dentistry, The Blizard Building, 4 Newark Street, London E1 2AT, UK
| | - Alvaro Mata
- School of Pharmacy, University of Nottingham, Nottingham NG7 2RD, UK
- Department of Chemical and Environmental Engineering, University of Nottingham, Nottingham NG7 2RD, UK
- Biodiscovery Institute, University of Nottingham, Nottingham, NG7 2RD, UK
| | - Clelia Dispenza
- Dipartimento di Ingegneria (DI), Università degli Studi di Palermo, Viale delle Scienze, Edificio 6, Palermo 90128, Italy
- Istituto di Biofisica (IBF), Consiglio Nazionale Delle Ricerche (CNR), Via U. La Malfa 153, Palermo 90146, Italy
| |
Collapse
|
33
|
Cal F, Sezgin Arslan T, Derkus B, Kiran F, Cengiz U, Arslan YE. Synthesis of Silica-Based Boron-Incorporated Collagen/Human Hair Keratin Hybrid Cryogels with the Potential Bone Formation Capability. ACS APPLIED BIO MATERIALS 2021; 4:7266-7279. [PMID: 35006956 DOI: 10.1021/acsabm.1c00805] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Tissue engineering and regenerative medicine have evolved into a different concept, the so-called clinical tissue engineering. Within this context, the synthesis of next-generation inorganic-organic hybrid constructs without the use of chemical crosslinkers emerges with a great potential for treating bone defects. Here, we propose a sophisticated approach for synthesizing cost-effective boron (B)- and silicon (Si)-incorporated collagen/hair keratin (B-Si-Col-HK) cryogels with the help of sol-gel reactions. In this approach, collagen and hair keratin were engaged with a B-Si network using tetraethyl orthosilicate as a silica precursor, and the obtained cryogels were characterized in depth with attenuated total reflectance-Fourier transform infrared spectroscopy, solid-state NMR, X-ray diffraction, thermogravimetric analysis, porosity and swelling tests, Brunauer-Emmett-Teller and Barrett-Joyner-Halenda analyses, frequency sweep and temperature-dependent rheology, contact angle analysis, micromechanical tests, and scanning electron microscopy with energy dispersive X-ray analysis. In addition, the cell survival and osteogenic features of the cryogels were evaluated by the MTS test, live/dead assay, immuno/histochemistry, and quantitative real-time polymerase chain reaction analyses. We conclude that the B-Si-networked Col-HK cryogels having good mechanical durability and osteoinductive features would have the potential bone formation capability.
Collapse
Affiliation(s)
- Fatma Cal
- Regenerative Biomaterials Laboratory, Department of Bioengineering, Faculty of Engineering, Canakkale Onsekiz Mart University, Canakkale 17100, Turkey
| | - Tugba Sezgin Arslan
- Personalized Medicine and Biosensing Research (PMBR) Laboratory, Chemistry Department, Faculty of Science, Ankara University, Ankara 06560, Turkey
| | - Burak Derkus
- Stem Cell Research Lab, Department of Chemistry, Faculty of Science, Ankara University, Ankara 06560, Turkey.,Interdisciplinary Research Unit for Advanced Materials (INTRAM), Department of Chemistry, Ankara University, Ankara 06560, Turkey
| | - Fadime Kiran
- Pharmabiotic Technologies Research Laboratory, Department of Biology, Faculty of Science, Ankara University, Ankara 06100, Turkey
| | - Ugur Cengiz
- Surface Science Research Laboratory, Department of Chemical Engineering, Engineering Faculty, Canakkale Onsekiz Mart University, Canakkale 17100, Turkey
| | - Yavuz Emre Arslan
- Regenerative Biomaterials Laboratory, Department of Bioengineering, Faculty of Engineering, Canakkale Onsekiz Mart University, Canakkale 17100, Turkey
| |
Collapse
|
34
|
Ozudogru E, Isik M, Eylem CC, Nemutlu E, Arslan YE, Derkus B. Decellularized spinal cord meninges extracellular matrix hydrogel that supports neurogenic differentiation and vascular structure formation. J Tissue Eng Regen Med 2021; 15:948-963. [PMID: 34463042 DOI: 10.1002/term.3240] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 08/17/2021] [Accepted: 08/24/2021] [Indexed: 01/09/2023]
Abstract
Decellularization of extracellular matrices offers an alternative source of regenerative biomaterials that preserve biochemical structure and matrix components of native tissues. In this study, decellularized bovine spinal cord meninges (dSCM)-derived extracellular matrix hydrogel (MeninGEL) is fabricated by employing a protocol that involves physical, chemical, and enzymatic processing of spinal meninges tissue and preserves the biochemical structure of meninges. The success of decellularization is characterized by measuring the contents of residual DNA, glycosaminoglycans, and hydroxyproline, while a proteomics analysis is applied to reveal the composition of MeninGEL. Frequency and temperature sweep rheometry show that dSCM forms self-supporting hydrogel at physiological temperature. The MeninGEL possesses excellent cytocompatibility. Moreover, it is evidenced with immuno/histochemistry and gene expression studies that the hydrogel induces growth-factor free differentiation of human mesenchymal stem cells into neural-lineage cells. Furthermore, MeninGEL instructs human umbilical vein endothelial cells to form vascular branching. With its innate bioactivity and low batch-to-batch variation property, the MeninGEL has the potential to be an off-the-shelf product in nerve tissue regeneration and restoration.
Collapse
Affiliation(s)
- Eren Ozudogru
- Regenerative Biomaterials Laboratory, Department of Bioengineering, Engineering Faculty, Canakkale Onsekiz Mart University, Canakkale, Turkey
| | - Melis Isik
- Stem Cell Research Lab, Department of Chemistry, Faculty of Science, Ankara University, Ankara, Turkey
| | - Cemil Can Eylem
- Analytical Chemistry Division, Faculty of Pharmacy, Hacettepe University, Ankara, Turkey
| | - Emirhan Nemutlu
- Analytical Chemistry Division, Faculty of Pharmacy, Hacettepe University, Ankara, Turkey.,Bioanalytic and Omics Laboratory, Faculty of Pharmacy, Hacettepe University, Ankara, Turkey
| | - Yavuz Emre Arslan
- Regenerative Biomaterials Laboratory, Department of Bioengineering, Engineering Faculty, Canakkale Onsekiz Mart University, Canakkale, Turkey
| | - Burak Derkus
- Stem Cell Research Lab, Department of Chemistry, Faculty of Science, Ankara University, Ankara, Turkey.,Interdisciplinary Research Unit for Advanced Materials (INTRAM), Ankara University, Ankara, Turkey
| |
Collapse
|
35
|
Zhang X, van Rijt S. 2D biointerfaces to study stem cell-ligand interactions. Acta Biomater 2021; 131:80-96. [PMID: 34237424 DOI: 10.1016/j.actbio.2021.06.044] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 06/18/2021] [Accepted: 06/28/2021] [Indexed: 02/07/2023]
Abstract
Stem cells have great potential in the field of tissue engineering and regenerative medicine due to their inherent regenerative capabilities. However, an ongoing challenge within their clinical translation is to elicit or predict the desired stem cell behavior once transplanted. Stem cell behavior and function are regulated by their interaction with biophysical and biochemical signals present in their natural environment (i.e., stem cell niches). To increase our understanding about the interplay between stem cells and their resident microenvironments, biointerfaces have been developed as tools to study how these substrates can affect stem cell behaviors. This article aims to review recent developments on fabricating cell-instructive interfaces to control cell adhesion processes towards directing stem cell behavior. After an introduction on stem cells and their natural environment, static surfaces exhibiting predefined biochemical signals to probe the effect of chemical features on stem cell behaviors are discussed. In the third section, we discuss more complex dynamic platforms able to display biochemical cues with spatiotemporal control using on-off ligand display, reversible ligand display, and ligand mobility. In the last part of the review, we provide the reader with an outlook on future designs of biointerfaces. STATEMENT OF SIGNIFICANCE: Stem cells have great potential as treatments for many degenerative disorders prevalent in our aging societies. However, an ongoing challenge within their clinical translation is to promote stem cell mediated regeneration once they are transplanted in the body. Stem cells reside within our bodies where their behavior and function are regulated by interactions with their natural environment called the stem cell niche. To increase our understanding about the interplay between stem cells and their niche, 2D materials have been developed as tools to study how specific signals can affect stem cell behaviors. This article aims to review recent developments on fabricating cell-instructive interfaces to control cell adhesion processes towards directing stem cell behavior.
Collapse
|
36
|
Macías I, Alcorta-Sevillano N, Infante A, Rodríguez CI. Cutting Edge Endogenous Promoting and Exogenous Driven Strategies for Bone Regeneration. Int J Mol Sci 2021; 22:7724. [PMID: 34299344 PMCID: PMC8306037 DOI: 10.3390/ijms22147724] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 07/14/2021] [Accepted: 07/15/2021] [Indexed: 12/11/2022] Open
Abstract
Bone damage leading to bone loss can arise from a wide range of causes, including those intrinsic to individuals such as infections or diseases with metabolic (diabetes), genetic (osteogenesis imperfecta), and/or age-related (osteoporosis) etiology, or extrinsic ones coming from external insults such as trauma or surgery. Although bone tissue has an intrinsic capacity of self-repair, large bone defects often require anabolic treatments targeting bone formation process and/or bone grafts, aiming to restore bone loss. The current bone surrogates used for clinical purposes are autologous, allogeneic, or xenogeneic bone grafts, which although effective imply a number of limitations: the need to remove bone from another location in the case of autologous transplants and the possibility of an immune rejection when using allogeneic or xenogeneic grafts. To overcome these limitations, cutting edge therapies for skeletal regeneration of bone defects are currently under extensive research with promising results; such as those boosting endogenous bone regeneration, by the stimulation of host cells, or the ones driven exogenously with scaffolds, biomolecules, and mesenchymal stem cells as key players of bone healing process.
Collapse
Affiliation(s)
- Iratxe Macías
- Stem Cells and Cell Therapy Laboratory, BioCruces Bizkaia Health Research Institute, Cruces University Hospital, Plaza de Cruces S/N, 48903 Barakaldo, Spain; (I.M.); (N.A.-S.)
| | - Natividad Alcorta-Sevillano
- Stem Cells and Cell Therapy Laboratory, BioCruces Bizkaia Health Research Institute, Cruces University Hospital, Plaza de Cruces S/N, 48903 Barakaldo, Spain; (I.M.); (N.A.-S.)
- University of Basque Country UPV/EHU, 48940 Leioa, Spain
| | - Arantza Infante
- Stem Cells and Cell Therapy Laboratory, BioCruces Bizkaia Health Research Institute, Cruces University Hospital, Plaza de Cruces S/N, 48903 Barakaldo, Spain; (I.M.); (N.A.-S.)
| | - Clara I. Rodríguez
- Stem Cells and Cell Therapy Laboratory, BioCruces Bizkaia Health Research Institute, Cruces University Hospital, Plaza de Cruces S/N, 48903 Barakaldo, Spain; (I.M.); (N.A.-S.)
| |
Collapse
|
37
|
Xie C, Ye J, Liang R, Yao X, Wu X, Koh Y, Wei W, Zhang X, Ouyang H. Advanced Strategies of Biomimetic Tissue-Engineered Grafts for Bone Regeneration. Adv Healthc Mater 2021; 10:e2100408. [PMID: 33949147 DOI: 10.1002/adhm.202100408] [Citation(s) in RCA: 66] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 04/16/2021] [Indexed: 12/21/2022]
Abstract
The failure to repair critical-sized bone defects often leads to incomplete regeneration or fracture non-union. Tissue-engineered grafts have been recognized as an alternative strategy for bone regeneration due to their potential to repair defects. To design a successful tissue-engineered graft requires the understanding of physicochemical optimization to mimic the composition and structure of native bone, as well as the biological strategies of mimicking the key biological elements during bone regeneration process. This review provides an overview of engineered graft-based strategies focusing on physicochemical properties of materials and graft structure optimization from macroscale to nanoscale to further boost bone regeneration, and it summarizes biological strategies which mainly focus on growth factors following bone regeneration pattern and stem cell-based strategies for more efficient repair. Finally, it discusses the current limitations of existing strategies upon bone repair and highlights a promising strategy for rapid bone regeneration.
Collapse
Affiliation(s)
- Chang Xie
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine and Department of Orthopedic Surgery of the Second Affiliated Hospital Zhejiang University School of Medicine Hangzhou 310058 China
- Zhejiang University‐University of Edinburgh Institute Zhejiang University School of Medicine and Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province Zhejiang University School of Medicine Hangzhou 314499 China
- Department of Sports Medicine Zhejiang University School of Medicine Hangzhou 310058 China
| | - Jinchun Ye
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine and Department of Orthopedic Surgery of the Second Affiliated Hospital Zhejiang University School of Medicine Hangzhou 310058 China
- Zhejiang University‐University of Edinburgh Institute Zhejiang University School of Medicine and Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province Zhejiang University School of Medicine Hangzhou 314499 China
| | - Renjie Liang
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine and Department of Orthopedic Surgery of the Second Affiliated Hospital Zhejiang University School of Medicine Hangzhou 310058 China
- Zhejiang University‐University of Edinburgh Institute Zhejiang University School of Medicine and Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province Zhejiang University School of Medicine Hangzhou 314499 China
| | - Xudong Yao
- The Fourth Affiliated Hospital Zhejiang University School of Medicine Yiwu 322000 China
| | - Xinyu Wu
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine and Department of Orthopedic Surgery of the Second Affiliated Hospital Zhejiang University School of Medicine Hangzhou 310058 China
- Zhejiang University‐University of Edinburgh Institute Zhejiang University School of Medicine and Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province Zhejiang University School of Medicine Hangzhou 314499 China
| | - Yiwen Koh
- Zhejiang University‐University of Edinburgh Institute Zhejiang University School of Medicine and Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province Zhejiang University School of Medicine Hangzhou 314499 China
| | - Wei Wei
- Zhejiang University‐University of Edinburgh Institute Zhejiang University School of Medicine and Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province Zhejiang University School of Medicine Hangzhou 314499 China
- China Orthopedic Regenerative Medicine Group (CORMed) Hangzhou 310058 China
| | - Xianzhu Zhang
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine and Department of Orthopedic Surgery of the Second Affiliated Hospital Zhejiang University School of Medicine Hangzhou 310058 China
- Zhejiang University‐University of Edinburgh Institute Zhejiang University School of Medicine and Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province Zhejiang University School of Medicine Hangzhou 314499 China
| | - Hongwei Ouyang
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine and Department of Orthopedic Surgery of the Second Affiliated Hospital Zhejiang University School of Medicine Hangzhou 310058 China
- Zhejiang University‐University of Edinburgh Institute Zhejiang University School of Medicine and Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province Zhejiang University School of Medicine Hangzhou 314499 China
- Department of Sports Medicine Zhejiang University School of Medicine Hangzhou 310058 China
- China Orthopedic Regenerative Medicine Group (CORMed) Hangzhou 310058 China
| |
Collapse
|
38
|
Alshehri S, Susapto HH, Hauser CAE. Scaffolds from Self-Assembling Tetrapeptides Support 3D Spreading, Osteogenic Differentiation, and Angiogenesis of Mesenchymal Stem Cells. Biomacromolecules 2021; 22:2094-2106. [PMID: 33908763 PMCID: PMC8382244 DOI: 10.1021/acs.biomac.1c00205] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 04/15/2021] [Indexed: 01/01/2023]
Abstract
The apparent rise of bone disorders demands advanced treatment protocols involving tissue engineering. Here, we describe self-assembling tetrapeptide scaffolds for the growth and osteogenic differentiation of human mesenchymal stem cells (hMSCs). The rationally designed peptides are synthetic amphiphilic self-assembling peptides composed of four amino acids that are nontoxic. These tetrapeptides can quickly solidify to nanofibrous hydrogels that resemble the extracellular matrix and provide a three-dimensional (3D) environment for cells with suitable mechanical properties. Furthermore, we can easily tune the stiffness of these peptide hydrogels by just increasing the peptide concentration, thus providing a wide range of peptide hydrogels with different stiffnesses for 3D cell culture applications. Since successful bone regeneration requires both osteogenesis and vascularization, our scaffold was found to be able to promote angiogenesis of human umbilical vein endothelial cells (HUVECs) in vitro. The results presented suggest that ultrashort peptide hydrogels are promising candidates for applications in bone tissue engineering.
Collapse
Affiliation(s)
- Salwa Alshehri
- Laboratory
for Nanomedicine, Division of Biological and Environmental
Science and Engineering and Computational Bioscience Research Center (CBRC), King Abdullah University of Science and Technology, Thuwal 23955-6900, Kingdom of Saudi Arabia
| | - Hepi H. Susapto
- Laboratory
for Nanomedicine, Division of Biological and Environmental
Science and Engineering and Computational Bioscience Research Center (CBRC), King Abdullah University of Science and Technology, Thuwal 23955-6900, Kingdom of Saudi Arabia
| | - Charlotte A. E. Hauser
- Laboratory
for Nanomedicine, Division of Biological and Environmental
Science and Engineering and Computational Bioscience Research Center (CBRC), King Abdullah University of Science and Technology, Thuwal 23955-6900, Kingdom of Saudi Arabia
| |
Collapse
|
39
|
Abbasi B, Harper J, Ahmadvand S. A short critique on biomining technology for critical materials. World J Microbiol Biotechnol 2021; 37:87. [PMID: 33881629 DOI: 10.1007/s11274-021-03048-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Accepted: 04/03/2021] [Indexed: 10/21/2022]
Abstract
Being around for several decades, there is a vast amount of academic research on biomining, and yet it contributes less to the mining industry compared to other conventional technologies. This critique briefly comments on the current status of biomining research, enumerates a number of primary challenges, and elaborates on some kinetically-oriented strategies and bottom-up policies to sustain biomining with focus on critical material extraction and rare earth elements (REEs). Finally, we present some edge cutting developments which may promote new potentials in biomining.
Collapse
Affiliation(s)
- Behrooz Abbasi
- Department of Mining and Metallurgical Engineering, University of Nevada, Reno, 89557, USA.
| | - Jeffrey Harper
- Department of Biochemistry and Molecular Biology, University of Nevada, Reno, 89557, USA.
| | | |
Collapse
|
40
|
Wu Y, Fortunato GM, Okesola BO, Brocchetti FLPD, Suntornnond R, Connelly J, De Maria C, Rodriguez-Cabello JC, Vozzi G, Wang W, Mata A. An interfacial self-assembling bioink for the manufacturing of capillary-like structures with tuneable and anisotropic permeability. Biofabrication 2021; 13. [PMID: 33561850 DOI: 10.1088/1758-5090/abe4c3] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 02/09/2021] [Indexed: 12/28/2022]
Abstract
Self-assembling bioinks offer the possibility to biofabricate with molecular precision, hierarchical control, and biofunctionality. For this to become a reality with widespread impact, it is essential to engineer these ink systems ensuring reproducibility and providing suitable standardization. We have reported a self-assembling bioink based on disorder-to-order transitions of an elastin-like recombinamer (ELR) to co-assemble with graphene oxide (GO). Here, we establish reproducible processes, optimize printing parameters for its use as a bioink, describe new advantages that the self-assembling bioink can provide, and demonstrate how to fabricate novel structures with physiological relevance. We fabricate capillary-like structures with resolutions down to ∼10µm in diameter and ∼2µm thick tube walls and use both experimental and finite element analysis to characterize the printing conditions, underlying interfacial diffusion-reaction mechanism of assembly, printing fidelity, and material porosity and permeability. We demonstrate the capacity to modulate the pore size and tune the permeability of the resulting structures with and without human umbilical vascular endothelial cells. Finally, the potential of the ELR-GO bioink to enable supramolecular fabrication of biomimetic structures was demonstrated by printing tubes exhibiting walls with progressively different structure and permeability.
Collapse
Affiliation(s)
- Yuanhao Wu
- School of Pharmacy, University of Nottingham, Nottingham NG7 2RD, United Kingdom.,Biodiscovery Institute, University of Nottingham, Nottingham NG7 2RD, United Kingdom.,Institute of Bioengineering, Queen Mary University of London, London E1 4NS, United Kingdom.,School of Engineering and Materials Science, Queen Mary University of London, London E1 4NS, United Kingdom
| | - Gabriele Maria Fortunato
- Research Center 'E. Piaggio' and Dipartimento di Ingegneria dell'Informazione, University of Pisa, Largo Lucio Lazzarino, Pisa 1-56122, Italy
| | - Babatunde O Okesola
- Institute of Bioengineering, Queen Mary University of London, London E1 4NS, United Kingdom.,School of Engineering and Materials Science, Queen Mary University of London, London E1 4NS, United Kingdom
| | | | - Ratima Suntornnond
- CREATE LAB, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London E1 2AT, United Kingdom
| | - John Connelly
- CREATE LAB, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London E1 2AT, United Kingdom
| | - Carmelo De Maria
- Research Center 'E. Piaggio' and Dipartimento di Ingegneria dell'Informazione, University of Pisa, Largo Lucio Lazzarino, Pisa 1-56122, Italy
| | | | - Giovanni Vozzi
- Research Center 'E. Piaggio' and Dipartimento di Ingegneria dell'Informazione, University of Pisa, Largo Lucio Lazzarino, Pisa 1-56122, Italy
| | - Wen Wang
- Institute of Bioengineering, Queen Mary University of London, London E1 4NS, United Kingdom.,School of Engineering and Materials Science, Queen Mary University of London, London E1 4NS, United Kingdom
| | - Alvaro Mata
- School of Pharmacy, University of Nottingham, Nottingham NG7 2RD, United Kingdom.,Biodiscovery Institute, University of Nottingham, Nottingham NG7 2RD, United Kingdom.,Institute of Bioengineering, Queen Mary University of London, London E1 4NS, United Kingdom.,School of Engineering and Materials Science, Queen Mary University of London, London E1 4NS, United Kingdom.,Department of Chemical and Environmental Engineering, University of Nottingham, Nottingham NG7 2RD, United Kingdom
| |
Collapse
|
41
|
Hu Q, Tang H, Yao Y, Liu S, Zhang H, Ramalingam M. Rapid fabrication of gelatin-based scaffolds with prevascularized channels for organ regeneration. Biomed Mater 2021; 16. [PMID: 33730706 DOI: 10.1088/1748-605x/abef7b] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Accepted: 03/17/2021] [Indexed: 12/13/2022]
Abstract
One of the biggest hinders in tissue engineering over the last decades was the complexity of the prevascularized channels of the engineered scaffold, which was still lower than that of human tissues. Another relative trouble was lacking precision molding capability, which restricted the clinical applications of the huge engineered scaffold. In this study, a promising approach was proposed to prepare hydrogel scaffold with prevascularized channels by liquid bath printing, which chitosan/β-sodium glycerophosphate (CS/β-GP) severed as the ink hydrogel, and gelation/nanoscale bacterial cellulose (Gel/BC) acted as the supporting hydrogel. Here, the ink hydrogel was printed by a versatile nozzle and embedded in the supporting hydrogel. Ink hydrogel transformed into liquid effluent at low temperature after cross-linking of gelatin by microbial transglutaminase (mTG). No residual template was seen on the channel surface after template removal. This preparation had a high degree of freedom in the geometry of the channel, which was demonstrated by making various prevascularized channels including circular, branched, and tree-shaped networks. The molding accuracy of the channel was detected by studying the roundness of the cross-section of the molded hollow channel, and the effect of the mechanical properties by adding BC to supporting hydrogel was analyzed. Human umbilical vein endothelial cells (HUVECs) were injected into the aforementioned channels and formed confluent and homogeneous distribution on the surface of channels. Altogether, these results showed that this approach can construct hydrogel scaffold with complex and accurate molding prevascularized channels, and had great potential to resolve urgent vascularization issue of bulk tissue-engineering scaffold.
Collapse
Affiliation(s)
- Qingxi Hu
- Shanghai University, 99, , Shanghai, 200444, CHINA
| | - Haihu Tang
- Shanghai University, 99, , Shanghai, 200444, CHINA
| | - Yuan Yao
- Shanghai University, 99, , Shanghai, 200444, CHINA
| | - Suihong Liu
- Rapid Manufacturing Engineering Center, Shanghai University, No.99 Shangda Road, BaoShan District, Shanghai, China, Shanghai, 200444, CHINA
| | | | - Murugan Ramalingam
- Vellore Institute of Technology, Vandalur - Kelambakkam Road, Chennai , Vellore, Tamil Nadu, 632014, INDIA
| |
Collapse
|
42
|
Charbonnier B, Hadida M, Marchat D. Additive manufacturing pertaining to bone: Hopes, reality and future challenges for clinical applications. Acta Biomater 2021; 121:1-28. [PMID: 33271354 DOI: 10.1016/j.actbio.2020.11.039] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 11/06/2020] [Accepted: 11/24/2020] [Indexed: 12/12/2022]
Abstract
For the past 20 years, the democratization of additive manufacturing (AM) technologies has made many of us dream of: low cost, waste-free, and on-demand production of functional parts; fully customized tools; designs limited by imagination only, etc. As every patient is unique, the potential of AM for the medical field is thought to be considerable: AM would allow the division of dedicated patient-specific healthcare solutions entirely adapted to the patients' clinical needs. Pertinently, this review offers an extensive overview of bone-related clinical applications of AM and ongoing research trends, from 3D anatomical models for patient and student education to ephemeral structures supporting and promoting bone regeneration. Today, AM has undoubtably improved patient care and should facilitate many more improvements in the near future. However, despite extensive research, AM-based strategies for bone regeneration remain the only bone-related field without compelling clinical proof of concept to date. This may be due to a lack of understanding of the biological mechanisms guiding and promoting bone formation and due to the traditional top-down strategies devised to solve clinical issues. Indeed, the integrated holistic approach recommended for the design of regenerative systems (i.e., fixation systems and scaffolds) has remained at the conceptual state. Challenged by these issues, a slower but incremental research dynamic has occurred for the last few years, and recent progress suggests notable improvement in the years to come, with in view the development of safe, robust and standardized patient-specific clinical solutions for the regeneration of large bone defects.
Collapse
|
43
|
Najdanović JG, Cvetković VJ, Stojanović ST, Vukelić-Nikolić MĐ, Živković JM, Najman SJ. Vascularization and osteogenesis in ectopically implanted bone tissue-engineered constructs with endothelial and osteogenic differentiated adipose-derived stem cells. World J Stem Cells 2021; 13:91-114. [PMID: 33584982 PMCID: PMC7859989 DOI: 10.4252/wjsc.v13.i1.91] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2020] [Revised: 11/01/2020] [Accepted: 11/17/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND A major problem in the healing of bone defects is insufficient or absent blood supply within the defect. To overcome this challenging problem, a plethora of approaches within bone tissue engineering have been developed recently. Bearing in mind that the interplay of various diffusible factors released by endothelial cells (ECs) and osteoblasts (OBs) have a pivotal role in bone growth and regeneration and that adjacent ECs and OBs also communicate directly through gap junctions, we set the focus on the simultaneous application of these cell types together with platelet-rich plasma (PRP) as a growth factor reservoir within ectopic bone tissue engineering constructs.
AIM To vascularize and examine osteogenesis in bone tissue engineering constructs enriched with PRP and adipose-derived stem cells (ASCs) induced into ECs and OBs.
METHODS ASCs isolated from adipose tissue, induced in vitro into ECs, OBs or just expanded were used for implant construction as followed: BPEO, endothelial and osteogenic differentiated ASCs with PRP and bone mineral matrix; BPUI, uninduced ASCs with PRP and bone mineral matrix; BC (control), only bone mineral matrix. At 1, 2, 4 and 8 wk after subcutaneous implantation in mice, implants were extracted and endothelial-related and bone-related gene expression were analyzed, while histological analyses were performed after 2 and 8 wk.
RESULTS The percentage of vascularization was significantly higher in BC compared to BPUI and BPEO constructs 2 and 8 wk after implantation. BC had the lowest endothelial-related gene expression, weaker osteocalcin immunoexpression and Spp1 expression compared to BPUI and BPEO. Endothelial-related gene expression and osteocalcin immunoexpression were higher in BPUI compared to BC and BPEO. BPEO had a higher percentage of vascularization compared to BPUI and the highest CD31 immunoexpression among examined constructs. Except Vwf, endothelial-related gene expression in BPEO had a later onset and was upregulated and well-balanced during in vivo incubation that induced late onset of Spp1 expression and pronounced osteocalcin immunoexpression at 2 and 8 wk. Tissue regression was noticed in BPEO constructs after 8 wk.
CONCLUSION Ectopically implanted BPEO constructs had a favorable impact on vascularization and osteogenesis, but tissue regression imposed the need for discovering a more optimal EC/OB ratio prior to considerations for clinical applications.
Collapse
Affiliation(s)
- Jelena G Najdanović
- Department of Biology and Human Genetics; Department for Cell and Tissue Engineering, Faculty of Medicine, University of Niš, Niš 18108, Serbia
| | - Vladimir J Cvetković
- Department of Biology and Ecology, Faculty of Sciences and Mathematics, University of Niš, Niš 18106, Serbia
| | - Sanja T Stojanović
- Department of Biology and Human Genetics; Department for Cell and Tissue Engineering, Faculty of Medicine, University of Niš, Niš 18108, Serbia
| | - Marija Đ Vukelić-Nikolić
- Department of Biology and Human Genetics; Scientific Research Center for Biomedicine; Faculty of Medicine, University of Niš, Niš 18108, Serbia
| | - Jelena M Živković
- Department of Biology and Human Genetics; Scientific Research Center for Biomedicine; Faculty of Medicine, University of Niš, Niš 18108, Serbia
| | - Stevo J Najman
- Department of Biology and Human Genetics; Department for Cell and Tissue Engineering, Faculty of Medicine, University of Niš, Niš 18108, Serbia
| |
Collapse
|
44
|
Derkus B. Human cardiomyocyte-derived exosomes induce cardiac gene expressions in mesenchymal stromal cells within 3D hyaluronic acid hydrogels and in dose-dependent manner. JOURNAL OF MATERIALS SCIENCE. MATERIALS IN MEDICINE 2021; 32:2. [PMID: 33469781 PMCID: PMC7815535 DOI: 10.1007/s10856-020-06474-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Accepted: 12/10/2020] [Indexed: 06/12/2023]
Abstract
Accomplishing a reliable lineage-specific differentiation of stem cells is vital in tissue engineering applications, however, this need remained unmet. Extracellular nanovesicles (particularly exosomes) have previously been shown to have this potential owing to their rich biochemical content including proteins, nucleic acids and metabolites. In this work, the potential of human cardiomyocytes-derived exosomes to induce in vitro cardiac gene expressions in human mesenchymal stem cells (hMSCs) was evaluated. Cardiac exosomes (CExo) were integrated with hyaluronic acid (HA) hydrogel, which was functionalized with tyramine (HA-Tyr) to enable the development of 3D (three dimensional), robust and bioactive hybrid cell culture construct through oxidative coupling. In HA-Tyr/CExo 3D hybrid hydrogels, hMSCs exhibited good viability and proliferation behaviours. Real time quantitative polymerase chain reaction (RT-qPCR) results demonstrated that cells incubated within HA-Tyr/CExo expressed early cardiac progenitor cell markers (GATA4, Nkx2.5 and Tbx5), but not cTnT, which is expressed in the late stages of cardiac differentiation and development. The expressions of cardiac genes were remarkably increased with increasing CExo concentration, signifying a dose-dependent induction of hMSCs. This report, to some extent, explains the potential of tissue-specific exosomes to induce lineage-specific differentiation. However, the strategy requires further mechanistic explanations so that it can be utilized in translational medicine.
Collapse
Affiliation(s)
- Burak Derkus
- Department of Chemistry, Faculty of Science, Ankara University, 06560, Ankara, Turkey.
| |
Collapse
|
45
|
Barrett DW, Okesola BO, Costa E, Thrasivoulou C, Becker DL, Mata A, Deprest JA, David AL, Chowdhury TT. Potential sealing and repair of human FM defects after trauma with peptide amphiphiles and Cx43 antisense. Prenat Diagn 2020; 41:89-99. [PMID: 33045764 DOI: 10.1002/pd.5826] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Revised: 08/01/2020] [Accepted: 09/04/2020] [Indexed: 12/23/2022]
Abstract
OBJECTIVE We examined whether peptide amphiphiles functionalised with adhesive, migratory or regenerative sequences could be combined with amniotic fluid (AF) to form plugs that repair fetal membrane (FM) defects after trauma and co-culture with connexin 43 (Cx43) antisense. METHODS We assessed interactions between peptide amphiphiles and AF and examined the plugs in FM defects after trauma and co-culture with the Cx43antisense. RESULTS Confocal microscopy confirmed directed self-assembly of peptide amphiphiles with AF to form a plug within minutes, with good mechanical properties. SEM of the plug revealed a multi-layered, nanofibrous network that sealed the FM defect after trauma. Co-culture of the FM defect with Cx43 antisense and plug increased collagen levels but reduced GAG. Culture of the FM defect with peptide amphiphiles incorporating regenerative sequences for 5 days, increased F-actin and nuclear cell contraction, migration and polarization of collagen fibers across the FM defect when compared to control specimens with minimal repair. CONCLUSIONS Whilst the nanoarchitecture revealed promising conditions to seal iatrogenic FM defects, the peptide amphiphiles need to be designed to maximize repair mechanisms and promote structural compliance with high mechanical tolerance that maintains tissue remodeling with Cx43 antisense for future treatment.
Collapse
Affiliation(s)
- David W Barrett
- Institute of Bioengineering, School of Engineering and Materials Science, Queen Mary University of London, London, UK
| | - Babatunde O Okesola
- Institute of Bioengineering, School of Engineering and Materials Science, Queen Mary University of London, London, UK
| | - Eleni Costa
- Institute of Bioengineering, School of Engineering and Materials Science, Queen Mary University of London, London, UK
| | | | - David L Becker
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Alvaro Mata
- Institute of Bioengineering, School of Engineering and Materials Science, Queen Mary University of London, London, UK.,Biodiscovery Institute, School of Pharmacy, Department of Chemical and Environmental Engineering, University of Nottingham, Nottingham, UK
| | - Jan A Deprest
- Department of Obstetrics and Gynecology, University Hospitals Leuven, Leuven, Belgium.,Institute for Women's Health, University College London, London, UK
| | - Anna L David
- Department of Obstetrics and Gynecology, University Hospitals Leuven, Leuven, Belgium.,Institute for Women's Health, University College London, London, UK.,NIHR University College London Hospitals Biomedical Research Centre, London, UK
| | - Tina T Chowdhury
- Institute of Bioengineering, School of Engineering and Materials Science, Queen Mary University of London, London, UK
| |
Collapse
|
46
|
Hedegaard CL, Mata A. Integrating self-assembly and biofabrication for the development of structures with enhanced complexity and hierarchical control. Biofabrication 2020; 12:032002. [DOI: 10.1088/1758-5090/ab84cb] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|